Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Gastroenterology ; 159(1): 227-240.e7, 2020 07.
Article in English | MEDLINE | ID: mdl-32179092

ABSTRACT

BACKGROUND & AIMS: A significant proportion of colorectal cancer (CRC) cases have familial aggregation but little is known about the genetic factors that contribute to these cases. We performed an exhaustive functional characterization of genetic variants associated with familial CRC. METHODS: We performed whole-exome sequencing analyses of 75 patients from 40 families with a history of CRC (including early-onset cases) of an unknown germline basis (discovery cohort). We also sequenced specific genes in DNA from an external replication cohort of 473 families, including 488 patients with colorectal tumors that had normal expression of mismatch repair proteins (validation cohort). We disrupted the Fas-associated factor 1 gene (FAF1) in DLD-1 CRC cells using CRISPR/Cas9 gene editing; some cells were transfected with plasmids that express FAF1 missense variants. Cells were analyzed by immunoblots, quantitative real-time polymerase chain reaction, and functional assays monitoring apoptosis, proliferation, and assays for Wnt signaling or nuclear factor (NF)-kappa-B activity. RESULTS: We identified predicted pathogenic variant in the FAF1 gene (c.1111G>A; p.Asp371Asn) in the discovery cohort; it was present in 4 patients of the same family. We identified a second variant in FAF1 in the validation cohort (c.254G>C; p.Arg85Pro). Both variants encoded unstable FAF1 proteins. Expression of these variants in CRC cells caused them to become resistant to apoptosis, accumulate beta-catenin in the cytoplasm, and translocate NF-kappa-B to the nucleus. CONCLUSIONS: In whole-exome sequencing analyses of patients from families with a history of CRC, we identified variants in FAF1 that associate with development of CRC. These variants encode unstable forms of FAF1 that increase resistance of CRC cells to apoptosis and increase activity of beta-catenin and NF-kappa-B.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Colorectal Neoplasms/genetics , Neoplastic Syndromes, Hereditary/genetics , Aged , Apoptosis/genetics , Cell Line, Tumor , Colorectal Neoplasms/pathology , DNA Mutational Analysis , Female , Gene Knockout Techniques , Genetic Predisposition to Disease , Germ-Line Mutation , Humans , Male , Middle Aged , Mutagenesis, Site-Directed , NF-kappa B/metabolism , Neoplastic Syndromes, Hereditary/pathology , Pedigree , Exome Sequencing , Wnt Signaling Pathway/genetics , beta Catenin/metabolism
2.
Int J Mol Sci ; 21(7)2020 Mar 30.
Article in English | MEDLINE | ID: mdl-32235485

ABSTRACT

Aceruloplasminemia is a rare autosomal recessive genetic disease characterized by mild microcytic anemia, diabetes, retinopathy, liver disease, and progressive neurological symptoms due to iron accumulation in pancreas, retina, liver, and brain. The disease is caused by mutations in the Ceruloplasmin (CP) gene that produce a strong reduction or absence of ceruloplasmin ferroxidase activity, leading to an impairment of iron metabolism. Most patients described so far are from Japan. Prompt diagnosis and therapy are crucial to prevent neurological complications since, once established, they are usually irreversible. Here, we describe the largest series of non-Japanese patients with aceruloplasminemia published so far, including 13 individuals from 11 families carrying 13 mutations in the CP gene (7 missense, 3 frameshifts, and 3 splicing mutations), 10 of which are novel. All missense mutations were studied by computational modeling. Clinical manifestations were heterogeneous, but anemia, often but not necessarily microcytic, was frequently the earliest one. This study confirms the clinical and genetic heterogeneity of aceruloplasminemia, a disease expected to be increasingly diagnosed in the Next-Generation Sequencing (NGS) era. Unexplained anemia with low transferrin saturation and high ferritin levels without inflammation should prompt the suspicion of aceruloplasminemia, which can be easily confirmed by low serum ceruloplasmin levels. Collaborative joint efforts are needed to better understand the pathophysiology of this potentially disabling disease.


Subject(s)
Ceruloplasmin/deficiency , Ceruloplasmin/genetics , Iron Metabolism Disorders/genetics , Neurodegenerative Diseases/genetics , Adult , Aged , Early Diagnosis , Female , Humans , Iron Metabolism Disorders/diagnosis , Iron Metabolism Disorders/pathology , Liver/pathology , Male , Middle Aged , Models, Molecular , Mutation , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/pathology
3.
Nucleic Acids Res ; 45(W1): W207-W214, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28521008

ABSTRACT

Integrative analysis of whole-genome/exome-sequencing data has been challenging, especially for the non-programming research community, as it requires simultaneously managing a large number of computational tools. Even computational biologists find it unexpectedly difficult to reproduce results from others or optimize their strategies in an end-to-end workflow. We introduce Germline Mutation Scoring Tool fOr Next-generation sEquencing data (GeMSTONE), a cloud-based variant prioritization tool with high-level customization and a comprehensive collection of bioinformatics tools and data libraries (http://gemstone.yulab.org/). GeMSTONE generates and readily accepts a shareable 'recipe' file for each run to either replicate previous results or analyze new data with identical parameters and provides a centralized workflow for prioritizing germline mutations in human disease within a streamlined workflow rather than a pool of program executions.


Subject(s)
Disease/genetics , Germ-Line Mutation , Software , Cloud Computing , Colorectal Neoplasms/genetics , High-Throughput Nucleotide Sequencing , Humans , Internet
4.
Genet Med ; 17(2): 131-42, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25058500

ABSTRACT

PURPOSE: Colorectal cancer is an important cause of mortality in the developed world. Hereditary forms are due to germ-line mutations in APC, MUTYH, and the mismatch repair genes, but many cases present familial aggregation but an unknown inherited cause. The hypothesis of rare high-penetrance mutations in new genes is a likely explanation for the underlying predisposition in some of these familial cases. METHODS: Exome sequencing was performed in 43 patients with colorectal cancer from 29 families with strong disease aggregation without mutations in known hereditary colorectal cancer genes. Data analysis selected only very rare variants (0-0.1%), producing a putative loss of function and located in genes with a role compatible with cancer. Variants in genes previously involved in hereditary colorectal cancer or nearby previous colorectal cancer genome-wide association study hits were also chosen. RESULTS: Twenty-eight final candidate variants were selected and validated by Sanger sequencing. Correct family segregation and somatic studies were used to categorize the most interesting variants in CDKN1B, XRCC4, EPHX1, NFKBIZ, SMARCA4, and BARD1. CONCLUSION: We identified new potential colorectal cancer predisposition variants in genes that have a role in cancer predisposition and are involved in DNA repair and the cell cycle, which supports their putative involvement in germ-line predisposition to this neoplasm.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms/genetics , Exome , Genetic Predisposition to Disease , Genetic Variation , High-Throughput Nucleotide Sequencing , Genetic Counseling , Germ-Line Mutation , Humans , Loss of Heterozygosity , Male , Pedigree , Reproducibility of Results
5.
Carcinogenesis ; 34(10): 2286-91, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23712746

ABSTRACT

Colorectal cancer (CRC) is the second leading cause of cancer-related death among men and women in Western countries. Once a tumour develops, a differentiated prognosis could be determined by lifestyle habits or inherited and somatic genetic factors. Finding such prognostic factors will be helpful in order to identify cases with a shorter survival or at a higher risk of recurrence that may benefit from more intensive treatment and follow-up surveillance. Sixteen CRC genetic susceptibility variants were directly genotyped in a cohort of 1235 CRC patients recruited by the EPICOLON Spanish consortium. Univariate Cox and multivariate regression analyses were performed taking as primary outcomes overall survival (OS), disease-free survival and recurrence-free interval. Genetic variants rs9929218 at 16q22.1 and rs10795668 at 10p14 may have an effect on OS. The G allele of rs9929218 was linked with a better OS [GG genotype, genotypic model: hazard ratio (HR) = 0.65, 95% confidence interval (CI) 0.45-0.93, P = 0.0179; GG/GA genotypes, dominant model: HR = 0.66, 95% CI 0.47-0.94, P = 0.0202]. Likewise, the G allele of rs10795668 was associated with better clinical outcome (GG genotype, genotypic model: HR = 0.73, 95% CI 0.53-1.01, P = 0.0570; GA genotype, genotypic model: HR = 0.66, 95% CI 0.47-0.92, P = 0.0137; GG/GA genotypes, dominant model: HR = 0.68, 95% CI 0.50-0.94, P = 0.0194). In conclusion, CRC susceptibility variants rs9929218 and rs10795668 may exert some influence in modulating patient's survival and they deserve to be further tested in additional CRC cohorts in order to confirm their potential as prognosis or predictive biomarkers.


Subject(s)
Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/genetics , Genetic Predisposition to Disease , Genetic Variation , Adult , Aged , Aged, 80 and over , Female , Follow-Up Studies , Genotype , Humans , Male , Middle Aged , Neoplasm Staging , Polymorphism, Single Nucleotide , Population Surveillance , Prognosis
6.
Cancer Prev Res (Phila) ; 14(2): 185-194, 2021 02.
Article in English | MEDLINE | ID: mdl-33115781

ABSTRACT

Familial colorectal cancer Type X (FCCTX) comprises a heterogeneous group of families with an increased risk of developing colorectal cancer and other related tumors, but with mismatch repair-proficient, microsatellite-stable (MSS) tumors. Unfortunately, the genetic basis underlying their cancer predisposition remains unknown. Although pathogenic germline variants in BRIP1 increase the risk of developing hereditary ovarian cancer, the involvement of BRIP1 in hereditary colorectal cancer is still not well known. In order to identify new BRIP1 variants associated with inherited colorectal cancer, affected and nonaffected individuals from 18 FCCTX or high-risk MSS colorectal cancer families were evaluated by whole-exome sequencing, and another 62 colorectal cancer patients from FCCTX or high-risk MSS colorectal cancer families were screened by a next-generation sequencing (NGS) multigene panel. The families were recruited at the Genetic Counseling Unit of Hospital Clínico San Carlos of Madrid. A total of three different BRIP1 mutations in three unrelated families were identified. Among them, there were two frameshift variants [c.1702_1703del, p.(Asn568TrpfsTer9) and c.903del, p.(Leu301PhefsTer2)] that result in the truncation of the protein and are thus classified as pathogenic (class 5). The remaining was a missense variant [c.2220G>T, p.(Gln740His)] considered a variant of uncertain significance (class 3). The segregation and loss-of-heterozygosity studies provide evidence linking the two BRIP1 frameshift variants to colorectal cancer risk, with suggestive but not definitive evidence that the third variant may be benign. The results here presented suggest that germline BRIP1 pathogenic variants could be associated with hereditary colorectal cancer predisposition.Prevention Relevance: We suggest that BRIP1 pathogenic germline variants may have a causal role in CRC as moderate cancer susceptibility alleles and be associated with hereditary CRC predisposition. A better understanding of hereditary CRC may provide important clues to disease predisposition and could contribute to molecular diagnostics, improved risk stratification, and targeted therapeutic strategies.


Subject(s)
Colorectal Neoplasms/genetics , Fanconi Anemia Complementation Group Proteins/genetics , Genetic Predisposition to Disease , Neoplastic Syndromes, Hereditary/genetics , RNA Helicases/genetics , Aged , Aged, 80 and over , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/pathology , DNA Mutational Analysis , Female , Frameshift Mutation , Humans , Male , Middle Aged , Neoplastic Syndromes, Hereditary/diagnosis , Neoplastic Syndromes, Hereditary/pathology , Pedigree , Exome Sequencing
7.
Cancers (Basel) ; 11(3)2019 03 13.
Article in English | MEDLINE | ID: mdl-30871259

ABSTRACT

Colorectal cancer (CRC) shows aggregation in some families but no alterations in the known hereditary CRC genes. We aimed to identify new candidate genes which are potentially involved in germline predisposition to familial CRC. An integrated analysis of germline and tumor whole-exome sequencing data was performed in 18 unrelated CRC families. Deleterious single nucleotide variants (SNV), short insertions and deletions (indels), copy number variants (CNVs) and loss of heterozygosity (LOH) were assessed as candidates for first germline or second somatic hits. Candidate tumor suppressor genes were selected when alterations were detected in both germline and somatic DNA, fulfilling Knudson's two-hit hypothesis. Somatic mutational profiling and signature analysis were also performed. A series of germline-somatic variant pairs were detected. In all cases, the first hit was presented as a rare SNV/indel, whereas the second hit was either a different SNV (3 genes) or LOH affecting the same gene (141 genes). BRCA2, BLM, ERCC2, RECQL, REV3L and RIF1 were among the most promising candidate genes for germline CRC predisposition. The identification of new candidate genes involved in familial CRC could be achieved by our integrated analysis. Further functional studies and replication in additional cohorts are required to confirm the selected candidates.

8.
Oncotarget ; 8(16): 26732-26743, 2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28423643

ABSTRACT

Germline mutations in POLE and POLD1 have been shown to cause predisposition to colorectal multiple polyposis and a wide range of neoplasms, early-onset colorectal cancer being the most prevalent. In order to find additional mutations affecting the proofreading activity of these polymerases, we sequenced its exonuclease domain in 155 patients with multiple polyps or an early-onset colorectal cancer phenotype without alterations in the known hereditary colorectal cancer genes. Interestingly, none of the previously reported mutations in POLE and POLD1 were found. On the other hand, among the genetic variants detected, only two of them stood out as putative pathogenic in the POLE gene, c.1359 + 46del71 and c.1420G > A (p.Val474Ile). The first variant, detected in two families, was not proven to alter correct RNA splicing. Contrarily, c.1420G > A (p.Val474Ile) was detected in one early-onset colorectal cancer patient and located right next to the exonuclease domain. The pathogenicity of this change was suggested by its rarity and bioinformatics predictions, and it was further indicated by functional assays in Schizosaccharomyces pombe. This is the first study to functionally analyze a POLE genetic variant outside the exonuclease domain and widens the spectrum of genetic changes in this DNA polymerase that could lead to colorectal cancer predisposition.


Subject(s)
Adenomatous Polyposis Coli/diagnosis , Adenomatous Polyposis Coli/genetics , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , DNA Polymerase III/genetics , DNA Polymerase II/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Adolescent , Adult , Age of Onset , Aged , Aged, 80 and over , Alleles , Amino Acid Substitution , Child , Child, Preschool , Colorectal Neoplasms/prevention & control , DNA Polymerase II/chemistry , DNA Polymerase III/chemistry , Female , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Testing , Humans , Infant , Infant, Newborn , Male , Middle Aged , Models, Molecular , Mutation , Pedigree , Poly-ADP-Ribose Binding Proteins/chemistry , Protein Conformation , Protein Domains/genetics , Young Adult
9.
PLoS One ; 11(4): e0153084, 2016.
Article in English | MEDLINE | ID: mdl-27078840

ABSTRACT

BACKGROUND: Common low-penetrance genetic variants have been consistently associated with colorectal cancer risk. AIM: To determine if these genetic variants are associated also with adenoma susceptibility and may improve selection of patients with increased risk for advanced adenomas and/or multiplicity (≥ 3 adenomas). METHODS: We selected 1,326 patients with increased risk for advanced adenomas and/or multiplicity and 1,252 controls with normal colonoscopy from population-based colorectal cancer screening programs. We conducted a case-control association study analyzing 30 colorectal cancer susceptibility variants in order to investigate the contribution of these variants to the development of subsequent advanced neoplasia and/or multiplicity. RESULTS: We found that 14 of the analyzed genetic variants showed a statistically significant association with advanced adenomas and/or multiplicity: the probability of developing these lesions increased with the number of risk alleles reaching a 2.3-fold risk increment in individuals with ≥ 17 risk alleles. CONCLUSIONS: Nearly half of the genetic variants associated with colorectal cancer risk are also related to advanced adenoma and/or multiplicity predisposition. Assessing the number of risk alleles in individuals within colorectal cancer screening programs may help to identify better a subgroup with increased risk for advanced neoplasia and/or multiplicity in the general population.


Subject(s)
Adenoma/genetics , Colorectal Neoplasms/genetics , Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide , Aged , Alleles , Case-Control Studies , Female , Gene Frequency , Genome-Wide Association Study , Genotype , Humans , Logistic Models , Male , Middle Aged , Multivariate Analysis , Risk Assessment/methods , Risk Assessment/statistics & numerical data , Risk Factors
10.
Eur J Hum Genet ; 24(10): 1501-5, 2016 10.
Article in English | MEDLINE | ID: mdl-27165003

ABSTRACT

Colorectal cancer (CRC) is one of the most common neoplasms in the world. Fanconi anemia (FA) is a very rare genetic disease causing bone marrow failure, congenital growth abnormalities and cancer predisposition. The comprehensive FA DNA damage repair pathway requires the collaboration of 53 proteins and it is necessary to restore genome integrity by efficiently repairing damaged DNA. A link between FA genes in breast and ovarian cancer germline predisposition has been previously suggested. We selected 74 CRC patients from 40 unrelated Spanish families with strong CRC aggregation compatible with an autosomal dominant pattern of inheritance and without mutations in known hereditary CRC genes and performed germline DNA whole-exome sequencing with the aim of finding new candidate germline predisposition variants. After sequencing and data analysis, variant prioritization selected only those very rare alterations, producing a putative loss of function and located in genes with a role compatible with cancer. We detected an enrichment for variants in FA DNA damage repair pathway genes in our familial CRC cohort as 6 families carried heterozygous, rare, potentially pathogenic variants located in BRCA2/FANCD1, BRIP1/FANCJ, FANCC, FANCE and REV3L/POLZ. In conclusion, the FA DNA damage repair pathway may play an important role in the inherited predisposition to CRC.


Subject(s)
Colorectal Neoplasms/genetics , DNA Repair/genetics , Fanconi Anemia/genetics , Genetic Predisposition to Disease , Germ-Line Mutation , BRCA1 Protein/genetics , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/pathology , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , Exome , Fanconi Anemia/epidemiology , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group E Protein/genetics , Female , Genes, Dominant , Humans , Male , Pedigree , Spain
11.
World J Gastroenterol ; 20(8): 1961-71, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24587672

ABSTRACT

Colorectal cancer (CRC) is one of the most frequent neoplasms and an important cause of mortality in the developed world. This cancer is caused by both genetic and environmental factors although 35% of the variation in CRC susceptibility involves inherited genetic differences. Mendelian syndromes account for about 5% of the total burden of CRC, with Lynch syndrome and familial adenomatous polyposis the most common forms. Excluding hereditary forms, there is an important fraction of CRC cases that present familial aggregation for the disease with an unknown germline genetic cause. CRC can be also considered as a complex disease taking into account the common disease-commom variant hypothesis with a polygenic model of inheritance where the genetic components of common complex diseases correspond mostly to variants of low/moderate effect. So far, 30 common, low-penetrance susceptibility variants have been identified for CRC. Recently, new sequencing technologies including exome- and whole-genome sequencing have permitted to add a new approach to facilitate the identification of new genes responsible for human disease predisposition. By using whole-genome sequencing, germline mutations in the POLE and POLD1 genes have been found to be responsible for a new form of CRC genetic predisposition called polymerase proofreading-associated polyposis.


Subject(s)
Colorectal Neoplasms/genetics , Genetic Predisposition to Disease , Adenomatous Polyposis Coli/diagnosis , Adenomatous Polyposis Coli/genetics , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Environment , Genetic Association Studies , Genetic Variation , Genome, Human , Geography , Germ-Line Mutation , Humans , Incidence , Polymorphism, Single Nucleotide , Risk Factors , Sequence Analysis, DNA
12.
PLoS One ; 9(4): e95022, 2014.
Article in English | MEDLINE | ID: mdl-24743384

ABSTRACT

Colorectal cancer is one of the most frequent neoplasms and an important cause of mortality in the developed world. Mendelian syndromes account for about 5% of the total burden of CRC, being Lynch syndrome and familial adenomatous polyposis the most common forms. Lynch syndrome tumors develop mainly as a consequence of defective DNA mismatch repair associated with germline mutations in MLH1, MSH2, MSH6 and PMS2. A significant proportion of variants identified by screening these genes correspond to missense or noncoding changes without a clear pathogenic consequence, and they are designated as "variants of uncertain significance", being the c.1852_1853delinsGC (p.K618A) variant in the MLH1 gene a clear example. The implication of this variant as a low-penetrance risk variant for CRC was assessed in the present study by performing a case-control study within a large cohort from the COGENT consortium-COST Action BM1206 including 18,723 individuals (8,055 colorectal cancer cases and 10,668 controls) and a case-only genotype-phenotype correlation with several clinical and pathological characteristics restricted to the Epicolon cohort. Our results showed no involvement of this variant as a low-penetrance variant for colorectal cancer genetic susceptibility and no association with any clinical and pathological characteristics including family history for this neoplasm or Lynch syndrome.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Colorectal Neoplasms/genetics , INDEL Mutation , Mutation, Missense , Nuclear Proteins/genetics , Adenosine Triphosphatases/genetics , Amino Acid Substitution , Cohort Studies , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , Female , Genetic Association Studies , Germ-Line Mutation , Humans , Male , Mismatch Repair Endonuclease PMS2 , MutL Protein Homolog 1 , MutS Homolog 2 Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL