Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Exp Mol Pathol ; 130: 104855, 2023 04.
Article in English | MEDLINE | ID: mdl-36736685

ABSTRACT

Detecting MLH1 promoter methylation is highly relevant to differentiate between possible Lynch syndrome patients or patients with sporadic causes of MLH1/PMS2 deficiency in colorectal (CRC) and endometrial cancers. Here, we aimed to develop a test for assessing MLH1 promoter methylation based in next generation sequencing (NGS), and to evaluate the concordance of MLH1 methylation and BRAF-V600 mutation status in CRC. For that, we performed a series of experiments with DNA from tumor, saliva and commercial control samples and our in house developed amplicon-based NGS test. In patients' samples, MLH1 methylation above 10% was only observed in tumors with MLH1/PMS2 loss. We confirmed the reproducibility and accuracy of MLH1 promoter analysis performing a serial dilution experiment with completely methylated and unmethylated control DNAs and a comparison between two NGS platforms (Ion Proton and Illumina). In MLH1/PMS2 deficient tumors, the MLH1 methylation status was concordant with the BRAF mutation status in 90% (18/20) of the cases. Our amplicon-based NGS test showed a great sensitivity and specificity for detecting MLH1 methylation in CRC samples, with a high agreement with the evaluation of BRAF mutation. This simple and affordable test could be used as a reflex test to identify patients with sporadic causes of MLH1/PMS2 deficiency in CRC, aiding to genetic test referral and identification of Lynch syndrome patients.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis , Colorectal Neoplasms , Humans , Proto-Oncogene Proteins B-raf/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Mismatch Repair Endonuclease PMS2/genetics , Reproducibility of Results , DNA Methylation/genetics , Mutation/genetics , MutL Protein Homolog 1/genetics , High-Throughput Nucleotide Sequencing , Microsatellite Instability , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Germ-Line Mutation
2.
Clin Transl Oncol ; 25(6): 1673-1681, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36586066

ABSTRACT

PURPOSE: Low expression of HER2 (HER2-low expression) in breast cancer (BC) has unique biological characteristics. However, whether HER2-low expression has an impact on neoadjuvant chemotherapy (NACT) in HER2-negative breast cancer remains unclear. METHODS: This study reviewed the clinicopathological data of patients with BC treated with NACT at a single hospital from January 2018 to July 2022. Baseline patient characteristics, efficacy of NACT, and survival data were compared between the HER2-0 and HER2-low groups. The impact of NACT on HER2 status also was investigated. Subgroup analyses based on hormone receptor (HR) status were performed to explore the impact of HR signaling on HER2 status during chemotherapy. RESULTS: The progesterone receptor-positive rate in the HER2-low group was significantly higher than that in HER2-0 group. The local treatment response of the HER2-low group was worse, but the disease-free survival rate of the HER2-low group was significantly better than that of the HER2-0 group. The proportion of patients with increased HER2 immunohistochemistry score after NACT was significantly higher in the HER2-0 group. Subgroup analysis showed that the efficacy of chemotherapy in HR + patients was significantly worse than in HR- patients, and HR + patients had a higher proportion of increased HER2 immunohistochemistry score after chemotherapy. Mechanistic studies suggested that MLH1 expression loss during chemotherapy might link HR signaling and regulation of HER2 expression. CONCLUSIONS: We found that HER2-low expressing BC exhibits differential sensitivity to chemotherapy compared to HER2-0 expressing BC. The regulation of HER2 expression by HR signaling may mediate aspects of chemoresistance.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Neoadjuvant Therapy , Receptor, ErbB-2/metabolism , Disease-Free Survival , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemotherapy, Adjuvant
3.
Int J Mol Sci ; 23(19)2022 Sep 30.
Article in English | MEDLINE | ID: mdl-36232851

ABSTRACT

Lynch syndrome (LS) is the main hereditary colorectal cancer syndrome. There have been few reports regarding the clinical and molecular characteristics of LS patients in Latin America; this is particularly true in the Mexican population, where no information is available. The present study aims to describe the clinical and molecular spectrum of variants in a cohort of patients diagnosed with LS in Mexico. We present a retrospective analysis of 412 patients with suspected LS, whose main site of cancer diagnosis was the colon (58.25%), followed by the endometrium (18.93%). Next-generation sequencing analysis, with an extensive multigene panel, showed that 27.1% (112/414) had a variant in one of the genes of the mismatch repair pathway (MMR); 30.4% (126/414) had a variant in non-MMR genes such as CHEK2, APC, MUTYH, BRCA1, and BRCA2; and 42.5% (176/414) had no genetic variants. Most of the variants were found in MLH1. Pathogenic variants (PVs) in MMR genes were identified in 65.7% (96/146) of the total PVs, and 34.24% (45/146) were in non-MMR genes. Molecular and clinical characterization of patients with LS in specific populations allowed personalized follow-up, with the option for targeted treatment with immune checkpoint inhibitors and the development of public health policies. Moreover, such characterization allows for family cascade testing and consequent prevention strategies.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/epidemiology , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Mismatch Repair/genetics , DNA-Binding Proteins/genetics , Female , Germ-Line Mutation , Humans , Immune Checkpoint Inhibitors , Mexico/epidemiology , MutS Homolog 2 Protein/genetics , Retrospective Studies
4.
Int J Mol Sci ; 22(5)2021 Feb 27.
Article in English | MEDLINE | ID: mdl-33673731

ABSTRACT

During meiosis, the number of crossovers vary in correlation to the length of prophase chromosome axes at the synaptonemal complex stage. It has been proposed that the regular spacing of the DNA loops, along with the close relationship of the recombination complexes and the meiotic axes are at the basis of this covariation. Here, we use a cytogenomic approach to investigate the relationship between the synaptonemal complex length and the DNA content in chicken oocytes during the pachytene stage of the first meiotic prophase. The synaptonemal complex to DNA ratios of specific chromosomes and chromosome segments were compared against the recombination rates obtained by MLH1 focus mapping. The present results show variations in the DNA packing ratios of macro- and microbivalents and also between regions within the same bivalent. Chromosome or chromosome regions with higher crossover rates form comparatively longer synaptonemal complexes than expected based on their DNA content. These observations are compatible with the formation of higher number of shorter DNA loops along meiotic axes in regions with higher recombination levels.


Subject(s)
Chromosomes, Mammalian/genetics , Crossing Over, Genetic , DNA/genetics , Meiosis , Oocytes/metabolism , Recombination, Genetic , Synaptonemal Complex , Animals , Chickens , DNA/chemistry , Female , MutL Protein Homolog 1/metabolism , Oocytes/cytology
5.
Gac Med Mex ; 157(6): 618-623, 2021.
Article in English | MEDLINE | ID: mdl-35108246

ABSTRACT

INTRODUCTION: Several genes determine the development of colorectal cancer (CRC), such as MLH1, which encodes a protein that participates in DNA repair. MLH1 hypermethylation has been associated with gene silencing. OBJECTIVE: To analyze the methylation of five regions of MLH1 CpG island in colorectal tumors from Mexican patients. MATERIALS AND METHODS: One hundred and one tumor tissue samples were obtained from Mexican patients with CRC who provided informed consent. DNA was subjected to bisulfite conversion. Methylation of all five regions of the CpG island was evaluated using methylation-specific PCR. RESULTS: The frequency of methylation in Mexican patients with CRC was 25%. Regions A and B methylation was the main observed pattern (60%). Female patients showed a higher frequency of methylation (71%; OR 3.085; CI: 1.85-8.03; p = 0.02), and out of total methylated samples, 80% corresponded to individuals older than 45 years (p < 0.05). CONCLUSION: We calculated a methylation frequency for the MLH1 gene of 25% in Mexican patients with CRC, with this being the first report for this population. Female patients and patients older than 45 years showed a higher frequency of methylation.


INTRODUCCIÓN: Varios genes determinan el desarrollo de cáncer colorrectal (CCR), como MLH1, el cual codifica una proteína que participa en la reparación del ADN. La hipermetilación de MLH1 ha sido asociado con silenciamiento génico. OBJETIVO: Analizar la metilación de cinco regiones de la isla CpG de MLH1 en tumores colorrectales de pacientes mexicanos. MATERIALES Y MÉTODOS: Se obtuvieron 101 muestras de tejido tumoral de pacientes mexicanos con CCR, quienes proporcionaron su consentimiento informado. El ADN fue sometido a conversión por bisulfito. La metilación de las cinco regiones de la isla CpG fue evaluada utilizando PCR específica para metilación. RESULTADOS: La frecuencia de metilación en pacientes mexicanos con CCR fue del 25%. La metilación de las regiones A y B fue el principal patrón observado (60%). Las pacientes de sexo femenino mostraron una mayor frecuencia de metilación (71%) (odds ratio: 3.085; intervalo de confianza; 1.85-8.03; p = 0.02); y del total de muestras metiladas, el 80% fueron individuos mayores de 45 años (p < 0.05). CONCLUSIÓN: Calculamos una frecuencia de metilación para el gen MLH1 del 25% en pacientes mexicanos con CCR, siendo el primer reporte para esta población. Pacientes de sexo femenino y pacientes mayores de 45 años mostraron una mayor frecuencia de metilación.


Subject(s)
Colorectal Neoplasms , DNA Methylation , MutL Protein Homolog 1 , Age Factors , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/genetics , CpG Islands , Female , Humans , Male , Mexico , Middle Aged , MutL Protein Homolog 1/genetics , Promoter Regions, Genetic , Sex Factors
6.
Fam Cancer ; 19(4): 323-336, 2020 10.
Article in English | MEDLINE | ID: mdl-32363481

ABSTRACT

Germline pathogenic variants in the DNA mismatch repair genes (MMR): MLH1, MSH2, MSH6, and PMS2, are causative of Lynch syndrome (LS). However, many of the variants mapping outside the invariant splice site positions (IVS ± 1, IVS ± 2) are classified as variants of unknown significance (VUS). Three such variants (MLH1 c.588+5G>C, c.588+5G>T and c.677+5G>A) were identified in 8 unrelated LS families from Argentina, Brazil and Chile. Herein, we collected clinical information on these families and performed segregation analysis and RNA splicing studies to assess the implication of these VUS in LS etiology. Pedigrees showed a clear pattern of variant co-segregation with colorectal cancer and/or other LS-associated malignancies. Tumors presented deficient expression of MLH1-PMS2 proteins in 7/7 of the LS families, and MSI-high status in 3/3 cases. Moreover, RNA analyses revealed that c.588+5G>C and c.588+5G>T induce skipping of exon 7 whereas c.677+5G>A causes skipping of exon 8. In sum, we report that the combined clinical findings in the families and the molecular studies provided the evidences needed to demonstrate that the three MLH1 variants are causative of LS and to classify c.588+5G>C and c.677+5G>A as class 5 (pathogenic), and c.588+5G>T as class 4 (likely-pathogenic). Our findings underline the importance of performing clinical and family analyses, as well as RNA splicing assays in order to determine the clinical significance of intronic variants, and contribute to the genetic counseling and clinical management of patients and their relatives.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Introns , MutL Protein Homolog 1/genetics , RNA Splice Sites , RNA Splicing , Adult , Argentina , Brazil , Chile , Colorectal Neoplasms/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism , DNA Mismatch Repair , Exons , Female , Genetic Counseling , Humans , Male , Middle Aged , Mismatch Repair Endonuclease PMS2/deficiency , Mismatch Repair Endonuclease PMS2/genetics , Mismatch Repair Endonuclease PMS2/metabolism , MutL Protein Homolog 1/deficiency , MutL Protein Homolog 1/metabolism , Pedigree , Protein Isoforms
7.
São Paulo; s.n; 2019. 148 p. ilust, tabelas, quadros.
Thesis in Portuguese | LILACS, Inca | ID: biblio-1049750

ABSTRACT

O câncer colorretal (CCR) é o terceiro tipo de câncer mais comum no mundo e a segunda causa de morte por câncer. Dados atuais apontam que o câncer de reto (CaRe) contribui para a maior incidência de CCR observada em pacientes jovens e é responsável por um aumento destes tumores em cerca de 75% nos últimos 40 anos. As variantes germinativas são reportadas em 14 a 16% dos indivíduos jovens com CCR, independente de história familiar de câncer. Ainda assim, a causa do desenvolvimento de CCR na maioria dos casos de indivíduos jovens ou famílias com múltiplos indivíduos afetados permanece desconhecida. Neste estudo, foram investigadas variantes germinativas em 76 pacientes com CaRe incluindo 29 classificados pelos critérios de Amsterdam I/II para a Síndrome de Lynch (Grupo 1) e 47 pacientes com idade igual ou inferior 40 anos (Grupo 2). O objetivo principal foi identificar variantes em 93 genes relacionados ao câncer que possam contribuir para o risco de desenvolvimento da doença usando sequenciamento de alto desempenho. Quinze variantes foram selecionadas e avaliadas em membros de oito famílias. Foram identificadas 153 variantes envolvendo 65 genes. Pacientes com câncer de reto apresentaram alta frequência de variantes germinativas em ATM (19%), APC (10%) e BRCA2 (9%). Variantes patogênicas ou likely-patogênicas foram identificadas em genes de alta (MSH2, MSH6, MLH1, MUTYH e BRCA2), moderada (ATM) e baixa (MTHFR e NOTCH1) penetrância, em 18% (14/76) dos pacientes, concordante com dados da literatura. Variantes características da Síndrome de Lynch foram identificadas em seis pacientes (6/76) e variantes associadas à polipose no gene MUTYH, em dois pacientes (2/76). Variantes patogênicas ou likely-patogênicas foram identificadas em 17% (8/47) dos pacientes jovens, especialmente em MUTYH (3/47), MTHFR (2/47), ATM, MSH6 e MLH1 (1/47 cada). Dentre estes, dois de 14 pacientes não reportaram câncer na família e seis tiveram pelo menos um caso de câncer na família, mas não preencheram os critérios clínicos para síndrome de predisposição hereditária ao câncer. Este estudo revelou 25 variantes novas (não identificadas em bancos de dados públicos ou reportadas em literatura). Em adição, foram observadas variantes germinativas em diferentes genes das vias MMR e de recombinação homóloga, incluindo ATM, BRCA2 e POLD1. Foram identificadas 15 variantes candidatas e ou associadas ao fenótipo nos genes ATM, APC, MSH2, MTHFR, CDKN2A, MUTYH e POLD1. Essas variantes foram avaliadas em 20 indivíduos de 8 famílias, sendo confirmadas nos probandos e identificadas em mais de um membro das famílias investigadas. A identificação de genes associados à predisposição ao CaRe tem potencial importância para o delineamento de estratégias mais eficientes de diagnóstico e aconselhamento genético em famílias com um alto risco de desenvolver este tumor (AU)


Colorectal cancer (CRC) is the third most common cancer worldwide and the second leading cause of cancer-related death. Recent studies suggest that rectal cancer (ReCa) contributes to the higher CRC incidence observed in young patients, and it is responsible for 75% of the increased incidence in colorectal tumors over the last 40 years. Germline variants have been reported in 14-16% of patients with early-onset CRC, regardless of family history of cancer. Nonetheless, the causes of CRC onset in most young patients or families with multiple affected cases remain unknown. We investigated germline variants in 76 ReCa patients, including 29 cases classified by the Amsterdam I/II criteria for Lynch Syndrome (Group 1) and 47 early-onset ReCa patients (≤40 years old, Group 2). The main objective was to identify variants in 93 cancer-related genes that can contribute to increased risk of the disease development using next-generation sequencing. Fifteen candidate variants were evaluated in eight selected families. Next-generation sequencing revealed 153 variants involving 65 genes. ReCa patients showed high frequency of germline variants in ATM (19%), APC (10%) and BRCA2 (9%). Pathogenic or likely-pathogenic variants were observed in high (MSH2, MSH6, MLH1, MUTYH, and BRCA2), moderate (ATM) and low (MTHFR and NOTCH1) penetrance genes, in 18% (14/76) of the patients, consistent with the literature data. Variants associated with Lynch Syndrome were identified in six patients (6/76), and variants involved in MUTYH-associated polyposis in two patients (2/76). Pathogenic or likely-pathogenic variants were identified in 17% (8/47) of early-onset patients, especially in the MUTYH (3/47), MTHFR (2/47), ATM, MSH6, and MLH1 (1/47 each) genes. Among these, two of fourteen patients had no family history of cancer and six reported at least one case of cancer in the family, but none of them met clinical criteria for the hereditary cancer syndrome. This study revealed 25 new variants (not reported in public databases or previous studies). In addition, germline variants were observed in several genes involved in MMR and homologous recombination (HR) pathways, including ATM, BRCA2 and POLD1. Fifteen candidates and associated to the phenotype variants were identified in the ATM, APC, MSH2, MTHFR, CDKN2A, MUTYH, and POLD1 genes. These variants were evaluated in 20 individuals (8 families), being confirmed in the index cases and identified in more than one relative of the evaluated families. The identification of genes associated with ReCa predisposition is crucial for outlining more efficient diagnostic strategies and for improving genetic counseling for families with high risk to develop this tumor type (AU)


Subject(s)
Humans , Male , Female , Middle Aged , DNA , Colorectal Neoplasms/genetics , Heredity , Early Detection of Cancer , Pathology, Molecular , High-Throughput Nucleotide Sequencing
8.
Genes Chromosomes Cancer ; 57(7): 350-358, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29520894

ABSTRACT

Inactivating mutations in the MLH1 gene cause the cancer predisposition Lynch syndrome, but for small coding genetic variants it is mostly unclear if they are inactivating or not. Nine such MLH1 variants have been identified in South American colorectal cancer (CRC) patients (p.Tyr97Asp, p.His112Gln, p.Pro141Ala, p.Arg265Pro, p.Asn338Ser, p.Ile501del, p.Arg575Lys, p.Lys618del, p.Leu676Pro), and evidence of pathogenicity or neutrality was not available for the majority of these variants. We therefore performed biochemical laboratory testing of the variant proteins and compared the results to protein in silico predictions on structure and conservation. Additionally, we collected all available clinical information of the families to come to a conclusion concerning their pathogenic potential and facilitate clinical diagnosis in the affected families. We provide evidence that four of the alterations are causative for Lynch syndrome, four are likely neutral and one shows compromised activity which can currently not be classified with respect to its pathogenic potential. The work demonstrates that biochemical testing, corroborated by congruent evolutionary and structural information, can serve to reliably classify uncertain variants when other data are insufficient.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Genetic Predisposition to Disease , MutL Protein Homolog 1/genetics , Mutation , Colorectal Neoplasms, Hereditary Nonpolyposis/ethnology , Computer Simulation , HEK293 Cells , Humans , Middle Aged , MutL Protein Homolog 1/chemistry , Protein Conformation , South America
9.
Biochimie ; 146: 87-96, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29175432

ABSTRACT

MLH1 and PMS2 proteins form the MutLα heterodimer, which plays a major role in DNA mismatch repair (MMR) in humans. Mutations in MMR-related proteins are associated with cancer, especially with colon cancer. The N-terminal region of MutLα comprises the N-termini of PMS2 and MLH1 and, similarly, the C-terminal region of MutLα is composed by the C-termini of PMS2 and MLH1, and the two are connected by linker region. The nuclear localization sequences (NLSs) necessary for the nuclear transport of the two proteins are found in this linker region. However, the exact NLS sequences have been controversial, with different sequences reported, particularly for MLH1. The individual components are not imported efficiently, presumably due to their C-termini masking their NLSs. In order to gain insights into the nuclear transport of these proteins, we solved the crystal structures of importin-α bound to peptides corresponding to the supposed NLSs of MLH1 and PMS2 and performed isothermal titration calorimetry to study their binding affinities. Both putative MLH1 and PMS2 NLSs can bind to importin-α as monopartite NLSs, which is in agreement with some previous studies. However, MLH1-NLS has the highest affinity measured by a natural NLS peptide, suggesting a major role of MLH1 protein in nuclear import compared to PMS2. Finally, the role of MLH1 and PMS2 in the nuclear transport of the MutLα heterodimer is discussed.


Subject(s)
Cell Nucleus/metabolism , DNA Mismatch Repair , Mismatch Repair Endonuclease PMS2/metabolism , MutL Protein Homolog 1/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Humans , Karyopherins/metabolism , Mice , Mismatch Repair Endonuclease PMS2/chemistry , Models, Molecular , MutL Protein Homolog 1/chemistry , Protein Conformation
11.
Medicina (B Aires) ; 76(3): 180-2, 2016.
Article in Spanish | MEDLINE | ID: mdl-27295708

ABSTRACT

Lynch syndrome is the most frequent syndrome in hereditary colorectal cancer, a family-specific deleterious mutations in genes encoding DNA reparation proteins: MLH1 (mutL homolog 1), MSH2, MSH6 (mutS homolog 2 y 6, respectively), PMS2 (PMS1 homolog 2, mismatch repair system component) y MUTYH (mutY DNA glycosylase). The c.2252_2253delAA, p.Lys751Serfs*3 mutation in MLH1 gene segregates with a haplotype reported in the northern region of Italy and whose origin was attributed to a founder effect. This mutation co-segregates with typical characteristics of Lynch syndrome, including early age at onset and multiple primary tumors in the same individual, a high frequency of pancreatic cancer, high microsatellite instability and lack of PMS2 expression. This report describes a mutation in an Argentinian patient with endometrioid adenocarcinoma of uterus. Her first-degree relatives had a history of colon cancer diagnosed before 50 years, fulfilling the Amsterdam Criteria I and Lynch syndrome II. The high pathogenicity associated to this mutation makes necessary the study of all members from families with hereditary cancer, allowing pre-symptomatic genetic diagnosis, early assessment and the instauration of preventive treatments.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Founder Effect , Mismatch Repair Endonuclease PMS2/genetics , MutL Protein Homolog 1/genetics , Mutation/genetics , DNA Repair/genetics , Female , Humans , Lynch Syndrome II/genetics , Middle Aged , Pedigree
12.
Medicina (B.Aires) ; Medicina (B.Aires);76(3): 180-182, June 2016. ilus, tab
Article in Spanish | LILACS | ID: biblio-841567

ABSTRACT

El síndrome de Lynch es la más frecuente de las neoplasias colorrectales hereditarias. Se origina por mutaciones germinales deletéreas familia-específicas en los genes que codifican proteínas de reparación del ADN: MLH1 (homólogo humano de mutL), MSH2 y MSH6 (homólogo humano de mutS 2 y 6, respectivamente), PMS2 (homólogo humano de PMS1 2) y MUTYH (homólogo humano de la ADN-glycosilasa mutY). La mutación c.2252_2253delAA, p.Lys751Serfs*3 en el exón 19 del gen MLH1 segrega con un haplotipo descripto en la región norte de Italia y cuyo origen fue atribuido a un efecto fundador. Esta mutación co-segrega con características típicas del síndrome de Lynch, incluyendo afectación temprana y múltiples tumores primarios en el mismo individuo, una alta frecuencia de cáncer pancreático, elevada inestabilidad microsatelital y falta de expresión de PMS2. En el presente trabajo se comunica dicha mutación en una paciente argentina con adenocarcinoma endometroide de útero en cuya historia familiar existen antecedentes de cáncer de colon diagnosticado antes de los 50 años en familiares de primer grado, reuniendo los criterios de Ámsterdam I y síndrome de Lynch II. Los polimorfismos presentes en la paciente coinciden con el haplotipo descripto en una región del norte de Italia. El alto grado de patogenicidad asociada a esta mutación hace imprescindible el estudio de todos los integrantes de las familias con cáncer hereditario permitiendo el diagnóstico genético pre-sintomático, la instauración de tratamientos o conductas preventivas y su seguimiento.


Lynch syndrome is the most frequent syndrome in hereditary colorectal cancer, a family-specific deleterious mutations in genes encoding DNA reparation proteins: MLH1 (mutL homolog 1), MSH2, MSH6 (mutS homolog 2 y 6, respectively), PMS2 (PMS1 homolog 2, mismatch repair system component) y MUTYH (mutY DNA glycosylase).The c.2252_2253delAA, p.Lys751Serfs*3 mutation in MLH1 gene segregates with a haplotype reported in the northern region of Italy and whose origin was attributed to a founder effect. This mutation co-segregates with typical characteristics of Lynch syndrome, including early age at onset and multiple primary tumors in the same individual, a high frequency of pancreatic cancer, high microsatellite instability and lack of PMS2 expression. This report describes a mutation in an Argentinian patient with endometrioid adenocarcinoma of uterus. Her first-degree relatives had a history of colon cancer diagnosed before 50 years, fulfilling the Amsterdam Criteria I and Lynch syndrome II. The high pathogenicity associated to this mutation makes necessary the study of all members from families with hereditary cancer, allowing pre-symptomatic genetic diagnosis, early assessment and the instauration of preventive treatments.


Subject(s)
Humans , Female , Middle Aged , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Founder Effect , Mismatch Repair Endonuclease PMS2/genetics , MutL Protein Homolog 1/genetics , Mutation/genetics , Pedigree , DNA Repair/genetics , Lynch Syndrome II/genetics
13.
Genetica ; 144(3): 307-12, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27115519

ABSTRACT

Meiotic recombination in female ducks was directly studied by immunolocalization of MLH1 protein, a mismatch repair protein of mature recombination nodules. In total, 6820 crossovers were scored along the autosomal synaptonemal complexes in 122 meiotic nuclei. From this analysis we predict that the female map length of the duck is 2845 cM, with a genome wide recombination rate of 2 cM/Mb. MLH1-focus mapping along the six largest bivalents shows regional variations of recombination frequencies that can be linked to differences in chromosome morphology. From this MLH1 mapping it can be inferred that distally located markers will appear more separated in genetic maps than physically equidistant markers located near the centromeres on bivalents 1 and 2. Instead, markers at interstitial positions on the acrocentric bivalents 3-6 will appear more tightly linked than expected on the basis of their physical distance because recombination is comparatively lower at the mid region of these chromosomes. The present results provide useful information to complement linkage mapping in ducks and extend previous knowledge about the variation of recombination rates among domestic Galloanserae.


Subject(s)
Ducks/genetics , Meiosis/genetics , Recombination, Genetic , Animals , Chromosome Mapping , Chromosomes , Crossing Over, Genetic , Female , Genetic Linkage , Genome , MutL Protein Homolog 1/genetics , Pachytene Stage/genetics , Synaptonemal Complex/genetics
14.
Arch Oral Biol ; 60(6): 825-33, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25791328

ABSTRACT

AIM: The aim of this study was to investigate the smoking habit influence on DNA methylation status in the promoters of the cancer related-genes MLH1, hTERT and TP53 in oral epithelial cells of healthy subjects. MATERIALS AND METHODS: DNA methylation analysis was performed using methylation-sensitive restriction enzymes (MSRE) in oral epithelial cells from non-smokers, smokers and ex-smokers. RESULTS: The investigated CpG dinucleotides located at HhaI and HpaII sites in the MLH1 gene promoter were observed to be fully methylated in the majority of DNA samples from the smoker group and statistical differences were found between non-smokers and smokers and between smokers and ex-smokers (p<0.05). The same was observed in the hTERT gene promoter at HhaI sites (p<0.05) and for HpaII sites the unmethylated condition was more frequent in smokers in comparison to non-smokers (p<0.05). For TP53, no differences were found among groups (p>0.05), with the fully methylated condition found to be a common event in healthy oral epithelial cells. CONCLUSION: We conclude that smoking may induce changes in DNA methylation status in cancer-related genes of oral epithelial cells and that the cessation of smoking is capable of reversing this process. Based on our data, we suggest that DNA methylation status of the hTERT and MLH1 gene promoters are promising markers for screening a set of smoking-related alterations in oral cells.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Biomarkers, Tumor/analysis , DNA Methylation , Epithelial Cells/pathology , Mouth Mucosa/cytology , Mouth Neoplasms/diagnosis , Mouth Neoplasms/genetics , Nuclear Proteins/genetics , Smoking , Tumor Suppressor Protein p53/genetics , Female , Genetic Markers , Humans , Male , Middle Aged , Mouth Mucosa/pathology , MutL Protein Homolog 1 , Polymerase Chain Reaction , Promoter Regions, Genetic
15.
Int J Cancer ; 137(4): 810-8, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25598504

ABSTRACT

We examined the influence of MLH1 c.-93G>A, MSH2 c.211 + 9C>G, MSH3 c.3133G>A and EXO1 c.1765G>A polymorphisms, involved in DNA mismatch repair (MMR), on head and neck (HN) squamous cell carcinoma (SCC) risk and prognosis. Aiming to identify genotypes, DNA from 450 HNSCC patients and 450 controls was analyzed by PCR-RFLP or real time PCR. MSH2 GG plus MSH3 GG (31.7% vs. 18.7%, p = 0.003) genotypes were higher in laryngeal SCC (LSCC) patients than in controls. Carriers of the respective combined genotype were under a 3.69 (95% CI: 1.54-8.81)-fold increased risk of LSCC. Interactions of tobacco and tobacco plus all the above-mentioned polymorphisms on HNSCC and LSCC risk were also evident in study (p = 0.001). At 60 months of follow-up, relapse-free survival (RFS) was shorter in patients with EXO1 GG genotype (54.8% vs. 61.1%, p = 0.03) and overall survival (OS) was shorter in patients with MSH3 GG genotype (42.8% vs. 52.5%, p = 0.02) compared to those with other genotypes, respectively. After multivariate Cox analysis, patients with EXO1 GG and MSH3 GG genotypes had worst RFS (HR: 1.50, 95% CI: 1.03-2.20, p = 0.03) and OS (HR: 1.59, 95% CI: 1.19-2.13, P = 0.002) than those with the remaining genotypes, respectively. Our data present, for the first time, evidence that inherited MLH1 c.-93G>A, MSH2 c.211 + 9C>G, MSH3 c.3133G>A, and EXO1 c.1765G>A abnormalities of DNA MMR pathway are important determinants of HNSCC, particularly among smokers, and predictors of patient outcomes.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carcinoma, Squamous Cell/genetics , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , Exodeoxyribonucleases/genetics , Head and Neck Neoplasms/genetics , MutS Homolog 2 Protein/genetics , Nuclear Proteins/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/pathology , Disease-Free Survival , Female , Genetic Association Studies , Genotype , Head and Neck Neoplasms/pathology , Humans , Male , Middle Aged , MutL Protein Homolog 1 , MutS Homolog 3 Protein , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Polymorphism, Single Nucleotide , Prognosis , Squamous Cell Carcinoma of Head and Neck
16.
World J Gastroenterol ; 19(20): 3043-51, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23716983

ABSTRACT

AIM: To evaluate the association between Helicobacter pylori (H. pylori) infection and MLH1 and MGMT methylation and its relationship with microsatellite instability (MSI). METHODS: The methylation status of the MLH1 and MGMT promoter region was analysed by methylation specific methylation-polymerase chain reaction (MSP-PCR) in gastric biopsy samples from uninfected or H. pylori-infected children (n = 50), from adults with chronic gastritis (n = 97) and from adults with gastric cancer (n = 92). MLH1 and MGMT mRNA expression were measured by real-time PCR and normalised to a constitutive gene (ß actin). MSI analysis was performed by screening MSI markers at 4 loci (Bat-25, Bat-26, D17S250 and D2S123) with PCR; PCR products were analysed by single strand conformation polymorphism followed by silver staining. Statistical analyses were performed with either the χ(2) test with Yates continuity correction or Fisher's exact test, and statistical significance for expression analysis was assessed using an unpaired Student's t-test. RESULTS: Methylation was not detected in the promoter regions of MLH1 and MGMT in gastric biopsy samples from children, regardless of H. pylori infection status. The MGMT promoter was methylated in 51% of chronic gastritis adult patients and was associated with H. pylori infection (P < 0.05); this region was methylated in 66% of gastric cancer patients, and the difference in the percentage of methylated samples between these patients and those from H. pylori-infected chronic gastritis patients was statistically significant (P < 0.05). MLH1 methylation frequencies among H. pylori-infected and non-infected chronic gastritis adult patients were 13% and 7%, respectively. We observed methylation of the MLH1 promoter (39%) and increased MSI levels (68%) in samples from gastric cancer patients in comparison to samples from H. pylori-infected adult chronic gastritis patients (P < 0.001 and P < 0.01, respectively). The frequency of promoter methylation for both genes was higher in gastric cancer samples than in H. pylori-positive chronic gastritis samples (P < 0.05). The levels of MLH1 and MGMT mRNA were significantly reduced in chronic gastritis samples that were also hypermethylated (P < 0.01). CONCLUSION: In summary, MGMT and MLH1 methylation did not occur in earlier-stage H. pylori infections and thus might depend on the duration of infection.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Gastritis/genetics , Helicobacter Infections/genetics , Helicobacter pylori/isolation & purification , Microsatellite Instability , Nuclear Proteins/genetics , Stomach Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Adolescent , Adult , Age Factors , Aged , Biopsy , Chi-Square Distribution , Child , Child, Preschool , Chronic Disease , Female , Gastritis/diagnosis , Gastritis/microbiology , Helicobacter Infections/diagnosis , Helicobacter Infections/microbiology , Humans , Male , Middle Aged , MutL Protein Homolog 1 , Promoter Regions, Genetic , Risk Factors , Stomach Neoplasms/diagnosis , Stomach Neoplasms/microbiology , Time Factors , Young Adult
17.
Campinas; s.n; maio 2013. 142 p. ilus, mapas, tab, graf.
Thesis in Portuguese | LILACS | ID: lil-706205

ABSTRACT

O câncer colorretal tem importância elevada frente a sua incidência e morbidade. Dentre os casos hereditários, o câncer colorretal hereditário sem polipose (HNPCC), ou Síndrome de Lynch, é responsável por cerca de 5% do total de casos. No HNPCC, a alteração genética herdada é a inativação de um dos alelos dos genes envolvidos em reparo do DNA, sendo os principais os genes hMLH1 e hMSH2. O objetivo deste trabalho foi investigar, em indivíduos com diagnóstico clínico de HNPCC, a presença de mutações nos genes MLH1 e MSH2, associar as variáveis clínicas com o gene mutado e investigar os familiares de portadores de HNPCC aos quais tivemos acesso, com relação a mutações germinativas. A investigação das mutações foi realizada por meio de sequenciamento direto dos éxons, região promotora e regiões de junção. Foram analisados 65 indivíduos divididos em três grupos, sendo (I) 46 pacientes portadores de câncer colorretal inclusos nos Critérios de Amsterdã, (II) dois familiares portadores de câncer colorretal e (III) 17 familiares sem câncer, todos da região metropolitana de Campinas, atendidos no Hospital de Clínicas da UNICAMP. Em 21 (45,65%) dos pacientes foram encontradas mutações deletérias. As mutações deletérias nos genes MLH1 e MSH2 estavam na proporção de 34,78% (16 pacientes) e 10,86% (5 pacientes), respectivamente. As mutações não deletérias nos genes MLH1 e MSH2 estavam na proporção de 65,22% dos pacientes (30 alterações) e 50% dos pacientes (23 alterações), respectivamente...


Colorectal cancer has high importance because of its incidence and morbidity. Among the hereditary cases, the hereditary nonpolyposis colorectal cancer (HNPCC) or Lynch syndrome, accounts for about 5% of cases. In HNPCC, the genetic alteration inherited is the inactivation of one of the alleles of genes involved in the DNA repair, being hMSH2 and hMLH1 the main genes. The objective of this study is to investigate the presence of mutations in MLH1 and MSH2 in patients with clinical diagnosis of HNPCC, correlate clinical variables with the mutated gene, and investigate the relatives of patients with HNPCC who we had access to, in relation to germline mutations. Investigation of the mutations was performed by éxons direct sequencing, the promoter and junction regions. Sixty-five individuals, divided into three groups, were studied: (I) 46 patients with colorectal cancer included in the Amsterdam Criteria, (II) two family members of colorectal cancer patients and (III) 17 relatives without cancer, all of them treated at Hospital das Clínicas at UNICAMP and living in the Campinas metropolitan area. Deleterious mutations were found in 21 patients (45.65%). The ratio of deleterious mutations in MLH1 and MSH2 was 34.78% (16 patients) and 10.86% (5 patients) respectively. The ratio of non deleterious mutations in genes MLH1 and MSH2 was 65.22% of patients (30 alterations) and 50% of patients (23 alterations) respectively. Among patients with HNPCC, 23 potentially deleterious mutations were identified, via sequences of MLH1 and MSH2 with a 50% detection rate. It doesn't seem to appear variations in the clinical characteristics of the tumor when a germline mutation occurs in MLH1 or MSH2, with the exception of the relationship between the presence of mutation in the MLH1 gene and age of disease onset. As it occurs throughout the world, the disease present a his molecular extremely heterogeneoty, where only two mutations were repeated in two patients...


Subject(s)
Humans , Male , Female , Young Adult , Middle Aged , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Clinical Diagnosis , Genes , Mutation
18.
Rev. méd. Chile ; 138(12): 1530-1534, dic. 2010. ilus
Article in Spanish | LILACS | ID: lil-583050

ABSTRACT

Background: About 30 percent of cases of colon cancer (CC) have a family history of CC, and only 5 percent are hereditary forms. Hereditary forms have an increased risk of CC and other tumors. Aim: To report the molecular and genetic study in two families with hereditary CC. Material and Methods: Molecular analysis of the adenomatous polyposis coli (APC) gene of familial adenomatous polyposis (FAP), was done in a patient with multiple benign polyps and his children. Molecular analysis was performed for MLH1 gene mutation of hereditary non-polyposis colon cancer (HNPCC) in an asymptomatic patient with family history of multiple cancers and his mother with a confrmed mutation in the MLH1 gene. Results: The patient with FAP had an insertion of 17 base pairs in exon 9 of the APC gene and two of his children had the same mutation. The patient with history of HNPCC did not have the family mutation on MLH1. Conclusions: In the case of FAP, molecular study was performed in his children since manifestations in carriers of the mutation may begin in childhood. If the second patient would have had the mutation, the study of his children could have been postponed until the age of 18, when the risk for CC is increased.


Subject(s)
Adult , Female , Humans , Male , Middle Aged , Adenomatous Polyposis Coli/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Pedigree , Chile , Mutation , Risk Factors
19.
Rev. méd. Chile ; 136(6): 757-762, jun. 2008. ilus
Article in Spanish | LILACS | ID: lil-490762

ABSTRACT

Hereditary non-polyposis colorectal cancer (HNPCC) or Lynch Syndrome is an autosomic dominant syndrome involving 596-1096 of colorectal cancer patients. Mutations in MLH1 and MSH2 genes account for most cases. These two genes particípate in the DNA mismatch repair pathway. Therefore mutation carriers show microsatellite instability (MSI) in tumors. This syndrome is characterized by the early development of colorectal cancer (before 50 years) and an increased incidence of cancer in other organs. We report four siblings from a family diagnosed with HNPCC. All of them were subjected to colonic surgery for colorectal cancer Moreover, one patient developed an ampulloma after her colon surgery. The molecular-genetic analysis revealed three brothers with microsatellite instability in the tumor tissue, the absence of the MLH1 protein, and the presence of a germ Une mutation localized in introm 15 ofthe MLH1 gene.


Subject(s)
Adult , Female , Humans , Male , Adenocarcinoma/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Mutation/genetics , Siblings , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Microsatellite Instability , Microsatellite Repeats , /genetics , /metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Pedigree
SELECTION OF CITATIONS
SEARCH DETAIL