Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.086
Filter
2.
Mol Nutr Food Res ; 68(10): e2300737, 2024 May.
Article in English | MEDLINE | ID: mdl-38700077

ABSTRACT

SCOPE: Yogurt consumption is related to a decreased risk of colorectal cancer (CRC), but whether such association is causal remains unclear. Patients with familial adenomatous polyposis (FAP) are at increased risk of CRC development. Here, the study investigates the efficacy of yogurt for intestinal polyposis chemoprevention in ApcMin/+ mice, a preclinical model for human FAP. METHODS AND RESULTS: A 10-week yogurt supplementation (15 g kg-1) in ApcMin/+ mice significantly reduces the intestinal polyp number (6.50 ± 0.97 versus 1.80 ± 0.49; p < 0.001) compared to controls. 16S rRNA gene-based microbiota analysis suggests that yogurt supplementation may greatly modulate the gut microbiome composition, especially in the relative abundance of Lactobacillus and Bifidobacterium. Importantly, the fecal concentration of d-lactate (d-Lac, 0.39 ± 0.04 µmol g-1 versus 8.14 ± 0.62 µmol g-1; p < 0.001) is boosted by yogurt, while oral administration with d-Lac (125 or 250 mg kg-1) reduces the polyp number by 71.43% or 77.14% (p < 0.001), respectively. The study also observes that d-Lac does not affect cell viability and anchorage-independence in CRC cells, but it greatly suppresses epidermal growth factor (EGF) or 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced cell transformation in preneoplastic cells. Mechanistically, it demonstrates that d-Lac may attenuate epithelial cell transformation by targeting PI3K/AKT/ß-catenin axis. CONCLUSION: Yogurt protects against intestinal polyposis in ApcMin/+ mice, and d-Lac may partially account for the chemopreventive effects above.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Yogurt , Animals , Colorectal Neoplasms/prevention & control , Gastrointestinal Microbiome/drug effects , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/prevention & control , Humans , Mice, Inbred C57BL , Mice , Male , Lactic Acid , Carcinogenesis/drug effects , Feces/microbiology , Feces/chemistry , Adenomatous Polyposis Coli Protein/genetics
3.
Br J Surg ; 111(5)2024 May 03.
Article in English | MEDLINE | ID: mdl-38722804

ABSTRACT

BACKGROUND: Hereditary adenomatous polyposis syndromes, including familial adenomatous polyposis and other rare adenomatous polyposis syndromes, increase the lifetime risk of colorectal and other cancers. METHODS: A team of 38 experts convened to update the 2008 European recommendations for the clinical management of patients with adenomatous polyposis syndromes. Additionally, other rare monogenic adenomatous polyposis syndromes were reviewed and added. Eighty-nine clinically relevant questions were answered after a systematic review of the existing literature with grading of the evidence according to Grading of Recommendations, Assessment, Development, and Evaluation methodology. Two levels of consensus were identified: consensus threshold (≥67% of voting guideline committee members voting either 'Strongly agree' or 'Agree' during the Delphi rounds) and high threshold (consensus ≥ 80%). RESULTS: One hundred and forty statements reached a high level of consensus concerning the management of hereditary adenomatous polyposis syndromes. CONCLUSION: These updated guidelines provide current, comprehensive, and evidence-based practical recommendations for the management of surveillance and treatment of familial adenomatous polyposis patients, encompassing additionally MUTYH-associated polyposis, gastric adenocarcinoma and proximal polyposis of the stomach and other recently identified polyposis syndromes based on pathogenic variants in other genes than APC or MUTYH. Due to the rarity of these diseases, patients should be managed at specialized centres.


Subject(s)
Adenocarcinoma , Adenomatous Polyposis Coli , DNA Glycosylases , Stomach Neoplasms , Humans , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/therapy , Adenomatous Polyposis Coli/diagnosis , Stomach Neoplasms/genetics , Stomach Neoplasms/therapy , Stomach Neoplasms/diagnosis , Adenocarcinoma/genetics , Adenocarcinoma/therapy , Adenocarcinoma/diagnosis , DNA Glycosylases/genetics , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/therapy , Neoplastic Syndromes, Hereditary/diagnosis , Europe , Adenomatous Polyps/genetics , Adenomatous Polyps/therapy , Polyps
4.
Wiad Lek ; 77(2): 338-344, 2024.
Article in English | MEDLINE | ID: mdl-38592998

ABSTRACT

OBJECTIVE: Aim: To explore the prevalence, clinical characteristics, and diagnostic aspects of diffuse familial adenomatous polyposis in childhood. This objective is accomplished through an extensive review of recent literature, and the presentation of case report from our clinical practice. PATIENTS AND METHODS: Materials and Methods: We analyzed 75 scientific papers, the findings of which have been documented in the PubMed database. Our search criteria included keywords such as ≪diffuse familial adenomatous intestinal polyposis,≫ ≪children,≫ and ≪diagnosis.≫ Then we conducted a second-stage analysis that involved a detailed review of a practical case - the medical records of inpatient Kh.V. who had been diagnosed with familial adenomatous polyposis. CONCLUSION: Conclusions: The analysis of the literature data is consistent with the findings from our clinical observations of familial adenomatous polyposis in a patient with complicated family anamnesis. It is worth noting that clinical features do not significantly differ across various types of polyposis. In cases of suspected familial adenomatous polyposis in adolescents, genetic testing is crucial.


Subject(s)
Adenomatous Polyposis Coli , Adolescent , Humans , Adenomatous Polyposis Coli/diagnosis , Adenomatous Polyposis Coli/complications , Adenomatous Polyposis Coli/genetics , Intestines , Genetic Testing
5.
JCO Precis Oncol ; 8: e2300404, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38564685

ABSTRACT

PURPOSE: Patients with germline pathogenic variants (PVs) in APC develop tens (attenuated familial adenomatous polyposis [AFAP]) to innumerable (classic FAP) adenomatous polyps in their colon and are at significantly increased lifetime risk of colorectal cancer. Up to 10% of FAP and up to 50% of patients with AFAP who have undergone DNA-only multigene panel testing (MGPT) do not have an identified PV in APC. We seek to demonstrate how the addition of RNA sequencing run concurrently with DNA can improve detection of germline PVs in individuals with a clinical presentation of AFAP/FAP. METHODS: We performed a retrospective query of individuals tested with paired DNA-RNA MGPT from 2021 to 2022 at a single laboratory and included those with a novel APC PV located in intronic regions infrequently covered by MGPT, a personal history of polyposis, and family medical history provided. All clinical data were deidentified in this institutional review board-exempt study. RESULTS: Three novel APC variants were identified in six families and were shown to cause aberrant splicing because of the creation of a deep intronic cryptic splice site that leads to an RNA transcript subject nonsense-mediated decay. Several carriers had previously undergone DNA-only genetic testing and had received a negative result. CONCLUSION: Here, we describe how paired DNA-RNA MGPT can be used to solve missing heritability in FAP families, which can have important implications in family planning and treatment decisions for patients and their families.


Subject(s)
Adenomatous Polyposis Coli , Colorectal Neoplasms , Humans , Retrospective Studies , Adenomatous Polyposis Coli/diagnosis , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Genetic Testing , Colorectal Neoplasms/genetics , DNA
6.
Cancer Sci ; 115(6): 1778-1790, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38566304

ABSTRACT

ABCC3 (also known as MRP3) is an ATP binding cassette transporter for bile acids, whose expression is downregulated in colorectal cancer through the Wnt/ß-catenin signaling pathway. However, it remained unclear how downregulation of ABCC3 expression contributes to colorectal carcinogenesis. We explored the role of ABCC3 in the progression of colorectal cancer-in particular, focusing on the regulation of bile acid export. Gene expression analysis of colorectal adenoma isolated from familial adenomatous polyposis patients revealed that genes related to bile acid secretion including ABCC3 were downregulated as early as at the stage of adenoma formation. Knockdown or overexpression of ABCC3 increased or decreased intracellular concentration of deoxycholic acid, a secondary bile acid, respectively, in colorectal cancer cells. Forced expression of ABCC3 suppressed deoxycholic acid-induced activation of MAPK signaling. Finally, we found that nonsteroidal anti-inflammatory drugs increased ABCC3 expression in colorectal cancer cells, suggesting that ABCC3 could be one of the targets for therapeutic intervention of familial adenomatous polyposis. Our data thus suggest that downregulation of ABCC3 expression contributes to colorectal carcinogenesis through the regulation of intracellular accumulation of bile acids and activity of MAPK signaling.


Subject(s)
Colorectal Neoplasms , Deoxycholic Acid , Down-Regulation , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , Multidrug Resistance-Associated Proteins , Humans , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Multidrug Resistance-Associated Proteins/metabolism , Multidrug Resistance-Associated Proteins/genetics , Deoxycholic Acid/pharmacology , Deoxycholic Acid/metabolism , Cell Line, Tumor , Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology
7.
Cancer Lett ; 589: 216822, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38521200

ABSTRACT

Familial adenomatous polyposis (FAP) is a heritable disease that increases the risk of colorectal cancer (CRC) development because of heterozygous mutations in APC. Little is known about the microenvironment of FAP. Here, single-cell RNA sequencing was performed on matched normal tissues, adenomas, and carcinomas from four patients with FAP. We analyzed the transcriptomes of 56,225 unsorted single cells, revealing the heterogeneity of each cell type, and compared gene expression among tissues. Then we compared the gene expression with that of sporadic CRC. Furthermore, we analyzed specimens of 26 FAP patients and 40 sporadic CRC patients by immunohistochemistry. Immunosuppressiveness of myeloid cells, fibroblasts, and regulatory T cells was upregulated even in the early stages of carcinogenesis. CD8+ T cells became exhausted only in carcinoma, although the cytotoxicity of CD8+ T cells was gradually increased according to the carcinogenic step. When compared with those in the sporadic CRC microenvironment, the composition and function of each cell type in the FAP-derived CRC microenvironment had differences. Our findings indicate that an immunosuppressive microenvironment is constructed from a precancerous stage in FAP.


Subject(s)
Adenoma , Adenomatous Polyposis Coli , Colorectal Neoplasms , Humans , CD8-Positive T-Lymphocytes/pathology , Adenomatous Polyposis Coli/complications , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Adenomatous Polyposis Coli Protein/genetics , Carcinogenesis , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Tumor Microenvironment
8.
Eur J Hum Genet ; 32(5): 588-592, 2024 May.
Article in English | MEDLINE | ID: mdl-38467732

ABSTRACT

In the Danish Polyposis Register, patients with over 100 cumulative colorectal adenomas of unknown genetic etiology, named in this study colorectal polyposis (CP), is registered and treated as familial adenomatous polyposis (FAP). In this study, we performed genetic analyses, including whole genome sequencing (WGS), of all Danish patients registered with CP and estimated the detection rate of pathogenic variants (PV). We identified 231 families in the Polyposis Register, 31 of which had CP. A polyposis-associated gene panel was performed and, if negative, patients were offered WGS and screening for mosaicism in blood and/or adenomas. Next-generation sequencing (NGS) was carried out for 27 of the families (four declined). PVs were detected in 11 families, and WGS revealed three additional structural variants in APC. Mosaicism of a PV in APC was detected in two families. As the variant detection rate of eligible families was 60%, 93% of families in the register now have a known genetic etiology.


Subject(s)
Adenomatous Polyposis Coli Protein , Adenomatous Polyposis Coli , Humans , Adenomatous Polyposis Coli/genetics , Female , Adenomatous Polyposis Coli Protein/genetics , Male , Denmark , Adult , Genotype , Middle Aged , Genetic Testing/methods , Mosaicism , Registries
9.
Mol Cancer Res ; 22(6): 515-523, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38546397

ABSTRACT

The pathogenesis of duodenal tumors in the inherited tumor syndromes familial adenomatous polyposis (FAP) and MUTYH-associated polyposis (MAP) is poorly understood. This study aimed to identify genes that are significantly mutated in these tumors and to explore the effects of these mutations. Whole exome and whole transcriptome sequencing identified recurrent somatic coding variants of phosphatidylinositol N-acetylglucosaminyltransferase subunit A (PIGA) in 19/70 (27%) FAP and MAP duodenal adenomas, and further confirmed the established driver roles for APC and KRAS. PIGA catalyzes the first step in glycosylphosphatidylinositol (GPI) anchor biosynthesis. Flow cytometry of PIGA-mutant adenoma-derived and CRISPR-edited duodenal organoids confirmed loss of GPI anchors in duodenal epithelial cells and transcriptional profiling of duodenal adenomas revealed transcriptional signatures associated with loss of PIGA. IMPLICATIONS: PIGA somatic mutation in duodenal tumors from patients with FAP and MAP and loss of membrane GPI-anchors may present new opportunities for understanding and intervention in duodenal tumorigenesis.


Subject(s)
Adenomatous Polyposis Coli , Duodenal Neoplasms , Glycosylphosphatidylinositols , Mutation , Humans , Glycosylphosphatidylinositols/metabolism , Glycosylphosphatidylinositols/genetics , Duodenal Neoplasms/genetics , Duodenal Neoplasms/metabolism , Duodenal Neoplasms/pathology , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Carcinogenesis/genetics , Male , Female
10.
Zhonghua Bing Li Xue Za Zhi ; 53(3): 224-229, 2024 Mar 08.
Article in Chinese | MEDLINE | ID: mdl-38433048

ABSTRACT

WHO firstly published the classification of paediatric tumours, in which genetic tumour syndromes were introduced as a separate chapter, covering the clinicopathological features, molecular genetic alterations, and diagnostic criteria of various tumor susceptibility syndromes common in children. This article briefly introduces and interprets 5 hotspot genetic tumour syndromes (neurofibromatosis type 1, naevoid basal cell carcinoma syndrome, von Hippel-Lindau syndrome, familial adenomatous polyposis and xeroderma pigmentosum) based on relevant literature, in order to bring new perspectives and insights to pathologists and clinicians.


Subject(s)
Adenomatous Polyposis Coli , Neoplasms , Child , Humans , Neoplasms/genetics , Adenomatous Polyposis Coli/genetics , Mutation , Pathologists , World Health Organization
11.
Gastroenterol Clin North Am ; 53(1): 179-200, 2024 03.
Article in English | MEDLINE | ID: mdl-38280747

ABSTRACT

Gastrointestinal polyposis disorders are a group of syndromes defined by clinicopathologic features that include the predominant histologic type of colorectal polyp and specific inherited gene mutations. Adenomatous polyposis syndromes comprise the prototypical familial adenomatous polyposis syndrome and other recently identified genetic conditions inherited in a dominant or recessive manner. Serrated polyposis syndrome is defined by arbitrary clinical criteria. The diagnosis of hamartomatous polyposis syndromes can be suggested from the histologic characteristics of colorectal polyps and the association with various extraintestinal manifestations. Proper identification of affected individuals is important due to an increased risk of gastrointestinal and extragastrointestinal cancers.


Subject(s)
Adenomatous Polyposis Coli , Colorectal Neoplasms , Humans , Colorectal Neoplasms/pathology , Adenomatous Polyposis Coli/diagnosis , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Syndrome
12.
J Gastroenterol ; 59(3): 187-194, 2024 03.
Article in English | MEDLINE | ID: mdl-38263336

ABSTRACT

BACKGROUND: Patients with familial adenomatous polyposis (FAP) have an increased risk of developing gastric neoplasms. However, the clinical course of FAP with these gastric lesions has not yet been fully clarified. The present study aimed to clarify the changes in the incidence risk of developing gastric adenoma or gastric cancer during the lifespan of patients with FAP. METHODS: Four hundred forty-three patients with data regarding gastric adenoma and gastric cancer retrospectively registered in a nationwide Japanese multicenter study were enrolled. The cumulative incidences and hazard rates (HRs) of gastric neoplasms were evaluated. RESULTS: The cumulative incidence rates in 50-year-old patients with FAP were 22.8% for gastric adenoma and 7.6% for gastric cancer, respectively. No significant association was found between gastric neoplasms and the colonic phenotype. The peak age for the HR of gastric adenoma was 65 years, with the highest HR (0.043). Regarding the incidence of gastric cancer, the HR increased moderately up to the age of 40 years, but the increase accelerated from the age of 50 years (HR = 0.0067). CONCLUSION: Careful surveillance of the upper gastrointestinal tract in elderly patients with FAP, such as shortening the interval of follow-up according to age, may be helpful for early diagnosis of gastric cancer.


Subject(s)
Adenocarcinoma , Adenomatous Polyposis Coli , Adenomatous Polyps , Stomach Neoplasms , Humans , Aged , Adult , Middle Aged , Stomach Neoplasms/etiology , Stomach Neoplasms/genetics , Japan/epidemiology , Adenomatous Polyposis Coli/complications , Adenomatous Polyposis Coli/epidemiology , Adenomatous Polyposis Coli/genetics , Adenocarcinoma/epidemiology , Adenocarcinoma/etiology , Adenocarcinoma/pathology
13.
Eur J Med Genet ; 68: 104913, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38286305

ABSTRACT

BACKGROUND: Familial Adenomatous Polyposis (FAP) is a colorectal cancer (CRC) predisposition syndrome caused by germline APC mutations and characterised by an increased risk of CRC and colonic polyps and, in certain forms, of specific prominent extraintestinal manifestations, namely osteomas, soft tissue tumours and dental anomalies. Pachydysostosis of the fibula is a rare clinical entity defined by unilateral bowing of the distal portion of the fibula and elongation of the entire bone, without affectation of the tibia. CLINICAL REPORT: We report a 17-year-old male, who presented with a non-progressive bowing of the right leg detected at 18 months of age caused by a fibula malformation (later characterized as pachydysostosis) and a large exophytic osteoma of the left radius, noticed at the age of 15 years, without gastrointestinal symptoms. There was no relevant family history. Detailed characterisation revealed multiple osteomas, skin lesions and dental abnormalities, raising the hypothesis of FAP. This diagnosis was confirmed by genetic testing [c.4406_4409dup p.(Ala1471Serfs*17) de novo mutation in the APC gene] and endoscopic investigation (multiple adenomas throughout the colon, ileum and stomach). DISCUSSION: This case report draws attention to the phenotypic spectrum of skeletal manifestations of FAP: this patient has a congenital fibula malformation, not previously associated with this syndrome, but which is likely to have been its first manifestation in this patient. This clinical case also illustrates the challenges in the early diagnosis of FAP, especially without family history, and highlights the importance of a multidisciplinary approach and the adequate study of rare skeletal abnormalities.


Subject(s)
Adenomatous Polyposis Coli , Osteoma , Male , Humans , Adolescent , Adenomatous Polyposis Coli Protein/genetics , Fibula/diagnostic imaging , Fibula/pathology , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/diagnosis , Genes, APC , Germ-Line Mutation , Osteoma/genetics
14.
Clin Transl Gastroenterol ; 15(1): e00645, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37856205

ABSTRACT

INTRODUCTION: Adenoma multiplicity is associated with increased colorectal cancer (CRC) risk. The utility of genetic testing in patients with multiple colorectal adenomas (MCRA) remains uncertain. We evaluated the diagnostic yield of mutations in polyposis- and CRC-associated genes in patients with MCRA. METHODS: We performed a cross-sectional review of adult patients with 10-99 cumulative adenomas from the prospective database at the St Mark's Hospital Polyposis Registry and Family Cancer Clinic between 1999 and 2021. Genetic testing was performed for adenomatous polyposis-associated genes, hamartomatous polyposis-associated genes, and nonpolyposis colorectal cancer-associated genes. Clinicopathological outcomes were extracted for multiple logistic regression analysis. RESULTS: Two hundred fifty-nine patients with MCRA (median age 61 [interquartile range 53-69] years) were identified. Sixty-six patients (25.5%) had a pathogenic variant or likely pathogenic variant, with APC and biallelic MUTYH mutations constituting the majority of identified pathogenic variant/likely pathogenic variants. Diagnostic yields were greater than 10% at any adenoma burden. In univariate analysis, higher adenoma burden and younger age were associated with higher yield (both P < 0.0001). In patients with MCRA with 10-19 adenomas without a relevant personal or family history of CRC, the diagnostic yield was nil. In multiple logistic regression analysis, higher adenoma burden, younger age, personal history of CRC, and first-degree familial history of CRC were associated with higher diagnostic yield. DISCUSSION: Diagnostic yield of >10% at any adenoma burden supports current guidance for constitutional genetic testing in patients with MCRA, although the low yield in people older than 60 years with 10-19 adenomas suggests that a stratified approach might be appropriate.


Subject(s)
Adenoma , Adenomatous Polyposis Coli , Colorectal Neoplasms , DNA Glycosylases , Adult , Humans , Middle Aged , Aged , Cohort Studies , Cross-Sectional Studies , DNA Glycosylases/genetics , Adenomatous Polyposis Coli/diagnosis , Adenomatous Polyposis Coli/genetics , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Genetic Testing , Adenoma/diagnosis , Adenoma/genetics , Adenoma/pathology
15.
Clin Genitourin Cancer ; 22(1): e31-e37.e1, 2024 02.
Article in English | MEDLINE | ID: mdl-37482523

ABSTRACT

INTRODUCTION: Somatic mutations in the Wnt signaling gene Adenomatous Polyposis Coli (APC) promote metastatic prostate cancer (PCa) progression. Less is known regarding the impact of germline APC mutations on PCa outcomes. We sought to investigate the prevalence of aggressive variant PCa (AVPC) and treatment-emergent neuroendocrine PCa (t-NEPC) in patients with the germline APC I1307K variant, an alteration found in 7% of Ashkenazi Jewish men. MATERIALS AND METHODS: We report a retrospective cohort study comparing patients with PCa and either APC I1307K germline mutation, APC somatic mutations, or unselected patients. Proportions of patients with AVPC among all the cases were estimated along with 95% Clopper-Pearson exact confidence intervals (CI). Odds ratios with 95% CI were provided for the prevalence of t-NEPC and AVPC in patients with germline APC I1307K compared to patients with frameshift alterations in APC. RESULTS: From 2016-2022, 18 patients with PCa at 3 institutions with the germline APC (I1307K) mutation were identified. Clinically-defined AVPC was found in 8 of the 15 cases with metastatic disease (53%; 95% CI: 26%-79%). Combined somatic alterations in two or more of RB1, TP53 or PTEN (molecularly-defined AVPC) were found in 5/18 cases (28%; 95% CI: 10%-54%). When compared to 20 patients with APC somatic frameshift mutations, patients with the germline APC I1307K variant had a significantly increased risk of AVPC (OR 7.2; 95% CI 1.27, 40.68). CONCLUSION: PCa that develops in the presence of the germline APC I1307K mutation appear to be enriched for clinically-defined and molecularly-defined AVPC and in particular, for t-NEPC.


Subject(s)
Adenomatous Polyposis Coli , Colorectal Neoplasms , Prostatic Neoplasms , Male , Humans , Germ-Line Mutation , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/genetics , Retrospective Studies , Adenomatous Polyposis Coli/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Germ Cells/pathology
16.
Gastroenterol Hepatol ; 47(4): 397-400, 2024 Apr.
Article in English, Spanish | MEDLINE | ID: mdl-37597744

ABSTRACT

Recently, biallelic MSH3 germline pathogenic/likely pathogenic variants have been recognized as a rare cause of adenomatous polyposis. We present a 49-year-old woman who was admitted to our high-risk colorectal cancer clinic after incidental detection of a biallelic MSH3 (likely) pathogenic variant when tested for the germline (likely) pathogenic variants in hereditary breast and ovarian cancer related genes. The focus of this case report is to describe the genotype and phenotype of our patient with MSH3-related adenomatous polyposis. More than half of the polyps (13/19) were located in the right colon. In addition, benign and malignant extraintestinal lesions may be common as our patient had simple liver and kidney cysts and two basal cell skin carcinomas.


Subject(s)
Adenomatous Polyposis Coli , Colonic Polyps , Colorectal Neoplasms , Female , Humans , Middle Aged , Colonic Polyps/genetics , Adenomatous Polyposis Coli/complications , Adenomatous Polyposis Coli/genetics , Genotype , Phenotype , Colorectal Neoplasms/genetics , MutS Homolog 3 Protein/genetics
17.
Intern Med ; 63(8): 1075-1079, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-37558482

ABSTRACT

Familial adenomatous polyposis (FAP) is caused by pathogenic variants of the APC gene on the long arm of chromosome 5. An analysis showed an association between germline APC gene variants and clinical signs of FAP; however, attenuated FAP has also been reported in cases with pathogenic variants. In contrast, a phenotype of FAP with no APC germline pathogenic variant and with few signs has been reported. We herein report a 16-year-old girl in whom the presence of multiple large bowel cancers from a young age and several small bowel cancers reflected a carcinogenic tendency higher than that typical for FAP.


Subject(s)
Adenomatous Polyposis Coli , Duodenal Neoplasms , Female , Humans , Adolescent , Adenomatous Polyposis Coli/genetics , Genes, APC , Germ-Line Mutation/genetics , Phenotype
18.
Genet Med ; 26(2): 100992, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37800450

ABSTRACT

PURPOSE: The Hereditary Colorectal Cancer/Polyposis Variant Curation Expert Panel (VCEP) was established by the International Society for Gastrointestinal Hereditary Tumours and the Clinical Genome Resource, who set out to develop recommendations for the interpretation of germline APC variants underlying Familial Adenomatous Polyposis, the most frequent hereditary polyposis syndrome. METHODS: Through a rigorous process of database analysis, literature review, and expert elicitation, the APC VCEP derived gene-specific modifications to the ACMG/AMP (American College of Medical Genetics and Genomics and Association for Molecular Pathology) variant classification guidelines and validated such criteria through the pilot classification of 58 variants. RESULTS: The APC-specific criteria represented gene- and disease-informed specifications, including a quantitative approach to allele frequency thresholds, a stepwise decision tool for truncating variants, and semiquantitative evaluations of experimental and clinical data. Using the APC-specific criteria, 47% (27/58) of pilot variants were reclassified including 14 previous variants of uncertain significance (VUS). CONCLUSION: The APC-specific ACMG/AMP criteria preserved the classification of well-characterized variants on ClinVar while substantially reducing the number of VUS by 56% (14/25). Moving forward, the APC VCEP will continue to interpret prioritized lists of VUS, the results of which will represent the most authoritative variant classification for widespread clinical use.


Subject(s)
Adenomatous Polyposis Coli , Genetic Testing , Humans , Genetic Testing/methods , Genetic Variation , Adenomatous Polyposis Coli/diagnosis , Adenomatous Polyposis Coli/genetics , Germ-Line Mutation/genetics , Germ Cells
19.
Gene ; 896: 148051, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38043837

ABSTRACT

Gardner syndrome (GS) is a specific form of familial adenomatous polyposis (FAP), which manifests as colorectal polyps, multiple osteomas and soft tissue tumors, and in the oral cavity as osteomas of the jaws, odontomas, and abnormal tooth counts. The underlying cause of GS is attributed to mutations in the APC gene. Mutations in this gene disrupt the normal functioning of the protein and lead to the development of GS. To further investigate GS, a family affected by the syndrome was selected from Dongguan, Guangdong Province. The family members underwent a comprehensive survey, which involved collecting clinical data and peripheral venous blood samples. The samples were then used for genetic analysis. Whole exome sequencing (WES) and Sanger sequencing techniques were utilized to screen and identify specific mutation sites in the APC gene. The clinical findings for the GS family included the presence of gastrointestinal polyps and odontomas. After analyzing the genetic sequencing results, a novel mutation site c.4266dupA on the APC gene was found in the patients, which leading to the APC protein truncation. As a result of this study, it is suggested that odontoma may be an early indicator of GS. Additionally, the identification of this novel mutation site in the APC gene expands the known spectrum of genetic mutations associated with the disease. This discovery has significant implications for the early diagnosis of GS, thus enabling timely intervention to reduce the risk of developing colon cancer and other related diseases.


Subject(s)
Adenomatous Polyposis Coli , Gardner Syndrome , Odontoma , Osteoma , Humans , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Adenomatous Polyposis Coli Protein/genetics , China , Gardner Syndrome/genetics , Gardner Syndrome/complications , Gardner Syndrome/pathology , Genes, APC , Germ-Line Mutation , Mutation , Odontoma/complications , Odontoma/genetics , Osteoma/complications , Osteoma/genetics
20.
Genes Chromosomes Cancer ; 63(1): e23208, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37795928

ABSTRACT

Polyketide synthase (pks) island harboring Escherichia coli are, under the right circumstances, able to produce the genotoxin colibactin. Colibactin is a risk factor for the development of colorectal cancer and associated with mutational signatures SBS88 and ID18. This study explores colibactin-associated mutational signatures in biallelic NTHL1 and MUTYH patients. Targeted Next Generation Sequencing (NGS) was performed on colorectal adenomas and carcinomas of one biallelic NTHL and 12 biallelic MUTYH patients. Additional fecal metagenomics and genome sequencing followed by mutational signature analysis was conducted for the NTHL1 patient. Targeted NGS of the NTHL1 patient showed somatic APC variants fitting SBS88 which was confirmed using WGS. Furthermore, fecal metagenomics revealed pks genes. Also, in 1 out of 11 MUTYH patient a somatic variant was detected fitting SBS88. This report shows that colibactin may influence development of colorectal neoplasms in predisposed patients.


Subject(s)
Adenomatous Polyposis Coli , Colorectal Neoplasms , Humans , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Mutation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Deoxyribonuclease (Pyrimidine Dimer)/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...