Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters

Publication year range
1.
Hum Genomics ; 18(1): 83, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39030589

ABSTRACT

BACKGROUND: Colorectal cancer is still the second leading cause of cancer-related deaths and thus biomarkers allowing prediction of the resistance of patients to therapy and estimating their prognosis are needed. We designed a panel of 558 genes with pharmacogenomics records related to 5-fluorouracil resistance, genes important for sensitivity to other frequently used drugs, major oncodrivers, and actionable genes. We performed a target enrichment sequencing of DNA from tumors and matched blood samples of patients, and compared the results with patient prognosis stratified by systemic adjuvant chemotherapy. RESULTS: The median number of detected variants per tumor sample was 18.5 with 4 classified as having a high predicted functional effect and 14.5 moderate effect. APC, TP53, and KRAS were the most frequent mutated genes (64%, 59%, and 42% of mutated samples, respectively) followed by FAT4 (23%), FBXW7, and PIK3CA (16% for both). Patients with advanced stage III had more frequently APC, TP53, or KRAS mutations than those in stages I or II. KRAS mutation counts followed an increasing trend with grade (G1 < G2 < G3). The response to adjuvant therapy was worse in carriers of frameshift mutations in APC or 12D variant in KRAS, but none of these oncodrivers had prognostic value. Carriage of somatic mutations in any of the genes ABCA13, ANK2, COL7A1, NAV3, or UNC80 had prognostic relevance for worse overall survival (OS) of all patients. In contrast, mutations in FLG, GLI3, or UNC80 were prognostic in the same direction for patients untreated, and mutations in COL6A3, LRP1B, NAV3, RYR1, RYR3, TCHH, or TENM4 for patients treated with adjuvant therapy. The first association was externally validated. From all germline variants with high or moderate predicted functional effects (median 326 per patient), > 5% frequency and positive Manhattan plot based on 3-year RFS, rs72753407 in NFACS, rs34621071 in ERBB4, and rs2444274 in RIF1 were significantly associated with RFS, OS or both. CONCLUSIONS: The present study identified several putative somatic and germline genetic events with prognostic potential for colorectal cancer that should undergo functional characterization.


Subject(s)
Colorectal Neoplasms , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Prognosis , Male , Female , Middle Aged , Aged , Mutation/genetics , Fluorouracil/therapeutic use , Biomarkers, Tumor/genetics , Adult , Drug Resistance, Neoplasm/genetics , Pharmacogenetics/methods , Adenomatous Polyposis Coli Protein/genetics , F-Box-WD Repeat-Containing Protein 7/genetics , Proto-Oncogene Proteins p21(ras)/genetics , High-Throughput Nucleotide Sequencing , Tumor Suppressor Protein p53/genetics , Class I Phosphatidylinositol 3-Kinases
2.
Cell Mol Life Sci ; 81(1): 57, 2024 Jan 27.
Article in English | MEDLINE | ID: mdl-38279052

ABSTRACT

The Wnt/ß-catenin pathway is critical to maintaining cell fate decisions. Recent study showed that liquid-liquid-phase separation (LLPS) of Axin organized the ß-catenin destruction complex condensates in a normal cellular state. Mutations inactivating the APC gene are found in approximately 80% of all human colorectal cancer (CRC). However, the molecular mechanism of the formation of ß-catenin destruction complex condensates organized by Axin phase separation and how APC mutations impact the condensates are still unclear. Here, we report that the ß-catenin destruction complex, which is constructed by Axin, was assembled condensates via a phase separation process in CRC cells. The key role of wild-type APC is to stabilize destruction complex condensates. Surprisingly, truncated APC did not affect the formation of condensates, and GSK 3ß and CK1α were unsuccessfully recruited, preventing ß-catenin phosphorylation and resulting in accumulation in the cytoplasm of CRCs. Besides, we propose that the phase separation ability of Axin participates in the nucleus translocation of ß-catenin and be incorporated and concentrated into transcriptional condensates, affecting the transcriptional activity of Wnt signaling pathway.


Subject(s)
Axin Signaling Complex , beta Catenin , Humans , Axin Signaling Complex/genetics , Axin Protein/genetics , Axin Protein/metabolism , beta Catenin/genetics , beta Catenin/metabolism , Phase Separation , Mutation/genetics , Wnt Signaling Pathway/genetics , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism
3.
Cancer ; 130(16): 2763-2769, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38630906

ABSTRACT

PURPOSE: Several recurring pathogenic variants (PVs) in BRCA1/BRCA2 and additional cancer susceptibility genes are described in the ethnically diverse Israeli population. Since 2019, testing for these recurring PVs is reimbursed unselectively for all patients with breast cancer (BC) in Israel. The aim was to evaluate the yield of genotyping for these PVs in non-Ashkenazi Jewish (AJ) patients with BC diagnosed ≥age 50 years. METHODS: Clinical and genotyping data of all patients with BC undergoing oncogenetic counseling at the Oncology Institute at Sheba Medical Center from June 2017 to December 2023 were reviewed. RESULTS: Of 2706 patients with BC (mean age at diagnosis, 54 years; range, 20-92 years) counseled, 515 patients of non-AJ (all four grandparents) descent, diagnosed ≥age 50 years of age were genotyped, 55 with triple-negative BC (TNBC) and 460 with non-TNBC. One of the recurring PVs in BRCA1/BRCA2 were detected in 12.7% (7/55) of TNBC patients and 0.65% (3/460) of non-TNBC. One patient with non-TNBC had PMS2 PV. Low-penetrance variants were found in 2.5% of genotyped TNBC and in 3.7% of patients with non-TNBC, including CHEK2 c.499G>A (n = 3), APC c.3920T > A (n = 4), and heterozygous MUTYH c.1187G>A (n = 5). Following first-pass genotyping, 146 patients performed multigene panel testing, none carried a BRCA1/BRCA2 PV, and only 5/127 non-TNBC (3.9%) harbored PVs in CHEK2 (n = 2, c.846+1G>C and c.592+3A>T), ATM c.103C>T (n = 2), and NBN c.966C>G (n = 1). CONCLUSIONS: The observed low rates of PV detection in non-AJ non-TNBC cases ≥age 50 years at diagnosis, mostly for clinically insignificant variants, questions the justification of unselected genotyping in this subset of patients.


Subject(s)
BRCA2 Protein , Breast Neoplasms , Checkpoint Kinase 2 , Genetic Predisposition to Disease , Jews , Humans , Middle Aged , Female , Aged , Adult , Checkpoint Kinase 2/genetics , Aged, 80 and over , Breast Neoplasms/genetics , Breast Neoplasms/ethnology , Jews/genetics , BRCA2 Protein/genetics , BRCA1 Protein/genetics , Israel/epidemiology , Genotype , Young Adult , Ataxia Telangiectasia Mutated Proteins/genetics , Mismatch Repair Endonuclease PMS2/genetics , Adenomatous Polyposis Coli Protein/genetics , DNA Glycosylases/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/ethnology , Genetic Testing/methods
4.
J Transl Med ; 22(1): 631, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38970018

ABSTRACT

BACKGROUND: Wnt/ß-catenin signalling impairment accounts for 85% of colorectal cancers (CRCs), including sporadic and familial adenomatous polyposis (FAP) settings. An altered PI3K/mTOR pathway and gut microbiota also contribute to CRC carcinogenesis. We studied the interplay between the two pathways and the microbiota composition within each step of CRC carcinogenesis. METHODS: Proteins and target genes of both pathways were analysed by RT-qPCR and IHC in tissues from healthy faecal immunochemical test positive (FIT+, n = 17), FAP (n = 17) and CRC (n = 15) subjects. CRC-related mutations were analysed through NGS and Sanger. Oral, faecal and mucosal microbiota was profiled by 16 S rRNA-sequencing. RESULTS: We found simultaneous hyperactivation of Wnt/ß-catenin and PI3K/mTOR pathways in FAP-lesions compared to CRCs. Wnt/ß-catenin molecular markers positively correlated with Clostridium_sensu_stricto_1 and negatively with Bacteroides in FAP faecal microbiota. Alistipes, Lachnospiraceae, and Ruminococcaceae were enriched in FAP stools and adenomas, the latter also showing an overabundance of Lachnoclostridium, which positively correlated with cMYC. In impaired-mTOR-mutated CRC tissues, p-S6R correlated with Fusobacterium and Dialister, the latter also confirmed in the faecal-ecosystem. CONCLUSIONS: Our study reveals an interplay between Wnt/ß-catenin and PI3K/mTOR, whose derangement correlates with specific microbiota signatures in FAP and CRC patients, and identifies new potential biomarkers and targets to improve CRC prevention, early adenoma detection and treatment.


Subject(s)
Carcinogenesis , Colorectal Neoplasms , Phosphatidylinositol 3-Kinases , TOR Serine-Threonine Kinases , Wnt Signaling Pathway , Humans , Colorectal Neoplasms/microbiology , TOR Serine-Threonine Kinases/metabolism , Pilot Projects , Phosphatidylinositol 3-Kinases/metabolism , Male , Female , Adenomatous Polyposis Coli/microbiology , Adenomatous Polyposis Coli/genetics , Middle Aged , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Feces/microbiology , Gastrointestinal Microbiome , Aged , Adult , Mutation/genetics , Microbiota
5.
Int J Mol Sci ; 25(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39125758

ABSTRACT

APC is a tumor suppressor gene that exerts its effect through the regulation of the Wnt signaling pathway. Loss of function mutations of the gene are associated with familial adenomatous polyposis (FAP). Early diagnosis in FAP patients is essential to prevent the development of colorectal cancer. Extraintestinal manifestations often precede the formation of the polyposis; therefore, these manifestations may serve as a clinical indicator for the condition. The aim of this study was to assess genotype-phenotype associations between the location of APC mutations and various extraintestinal features, mainly focusing on osseous and dental anomalies. Analyses of our cases and the mutations available in the literature with these manifestations revealed that mutations in the N-terminal region (amino acids 1-~1000) of the protein are more frequently associated with only osseous anomalies, whereas dental manifestations are more prevalent in mutations in the middle region (amino acids 1000-~2100). In addition, supernumerary teeth were found to be the most common dental feature. Since dental abnormalities often precede intestinal polyposis, dentists have a crucial role in the early identification of patients at risk.


Subject(s)
Adenomatous Polyposis Coli Protein , Adenomatous Polyposis Coli , Germ-Line Mutation , Humans , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli/genetics , Tooth Abnormalities/genetics , Genetic Association Studies , Tooth, Supernumerary/genetics , Genetic Predisposition to Disease , Male , Female
6.
Int J Mol Sci ; 25(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38891848

ABSTRACT

DNA methylation is an epigenetic process that commonly occurs in genes' promoters and results in the transcriptional silencing of genes. DNA methylation is a frequent event in bladder cancer, participating in tumor initiation and progression. Bladder cancer is a major health issue in patients suffering from neurogenic lower urinary tract dysfunction (NLUTD), although the pathogenetic mechanisms of the disease remain unclear. In this population, bladder cancer is characterized by aggressive histopathology, advanced stage during diagnosis, and high mortality rates. To assess the DNA methylation profiles of five genes' promoters previously known to be associated with bladder cancer in bladder tissue of NLUTD patients, we conducted a prospective study recruiting NLUTD patients from the neuro-urology unit of a public teaching hospital. Cystoscopy combined with biopsy for bladder cancer screening was performed in all patients following written informed consent being obtained. Quantitative methylation-specific PCR was used to determine the methylation status of RASSF1, RARß, DAPK, hTERT, and APC genes' promoters in bladder tissue samples. Twenty-four patients suffering from mixed NLUTD etiology for a median duration of 10 (IQR: 12) years were recruited in this study. DNA hypermethylation was detected in at least one gene of the panel in all tissue samples. RAR-ß was hypermethylated in 91.7% samples, RASSF and DAPK were hypermethylated in 83.3% samples, APC 37.5% samples, and TERT in none of the tissue samples. In 45.8% of the samples, three genes of the panel were hypermethylated, in 29.2% four genes were hypermethylated, and in 16.7% and in 8.3% of the samples, two and one gene were hypermethylated, respectively. The number of hypermethylated genes of the panel was significantly associated with recurrent UTIs (p = 0.0048). No other significant association was found between DNA hypermethylation or the number of hypermethylated genes and the clinical characteristics of the patients. Histopathological findings were normal in 8.3% of patients, while chronic inflammation was found in 83.3% of patients and squamous cell metaplasia in 16.7% of patients. In this study, we observed high rates of DNA hypermethylation of genes associated with bladder cancer in NLUTD patients, suggesting an epigenetic field effect and possible risk of bladder cancer development. Recurrent UTIs seem to be associated with increased DNA hypermethylation. Further research is needed to evaluate the impact of recurrent UTIs and chronic inflammation in DNA hypermethylation and bladder cancer etiopathogenesis in NLUTD patients.


Subject(s)
DNA Methylation , Promoter Regions, Genetic , Urinary Bladder Neoplasms , Humans , DNA Methylation/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Male , Female , Promoter Regions, Genetic/genetics , Middle Aged , Aged , Urinary Bladder/pathology , Prospective Studies , Tumor Suppressor Proteins/genetics , Urinary Bladder, Neurogenic/genetics , Epigenesis, Genetic , Telomerase/genetics , Death-Associated Protein Kinases/genetics , Adenomatous Polyposis Coli Protein/genetics , Receptors, Retinoic Acid
7.
Int J Mol Sci ; 25(10)2024 May 14.
Article in English | MEDLINE | ID: mdl-38791382

ABSTRACT

The identification of targets that are expressed on the cell membrane is a main goal in cancer research. The Lymphocyte Antigen 6 Family Member G6D (LY6G6D) gene codes for a protein that is mainly present on the surface of colorectal cancer (CRC) cells. Therapeutic strategies against this protein like the development of T cell engagers (TCE) are currently in the early clinical stage. In the present work, we interrogated public genomic datasets including TCGA to evaluate the genomic and immunologic cell profile present in tumors with high expression of LY6G6D. We used data from TCGA, among others, and the Tumor Immune Estimation Resource (TIMER2.0) platform for immune cell estimations and Spearman correlation tests. LY6G6D expression was exclusively present in CRC, particularly in the microsatellite stable (MSS) subtype, and was associated with left-side tumors and the canonical genomic subgroup. Tumors with mutations of APC and p53 expressed elevated levels of LY6G6D. This protein was expressed in tumors with an inert immune microenvironment with an absence of immune cells and co-inhibitory molecules. In conclusion, we described clinical, genomic and immune-pathologic characteristics that can be used to optimize the clinical development of agents against this target. Future studies should be performed to confirm these findings and potentially explore the suggested clinical development options.


Subject(s)
Colorectal Neoplasms , Colorectal Neoplasms/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Humans , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Female , Male , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Gene Expression Regulation, Neoplastic , Mutation , Middle Aged , Aged , Biomarkers, Tumor/genetics , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Antigens, Ly/metabolism , Antigens, Ly/genetics , B7 Antigens/genetics , B7 Antigens/metabolism
8.
Int J Mol Sci ; 25(12)2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38928449

ABSTRACT

Triple-negative breast cancer (TNBC) patients are treated with traditional chemotherapy, such as the taxane class of drugs. One such drug, paclitaxel (PTX), can be effective in treating TNBC; however, many tumors will develop drug resistance, which can lead to recurrence. In order to improve patient outcomes and survival, there lies a critical need to understand the mechanism behind drug resistance. Our lab made the novel observation that decreased expression of the Adenomatous Polyposis Coli (APC) tumor suppressor using shRNA caused PTX resistance in the human TNBC cell line MDA-MB-157. In cells lacking APC, induction of apoptosis by PTX was decreased, which was measured through cleaved caspase 3 and annexin/PI staining. The current study demonstrates that CRISPR-mediated APC knockout in two other TNBC lines, MDA-MB-231 and SUM159, leads to PTX resistance. In addition, the cellular consequences and molecular mechanisms behind APC-mediated PTX response have been investigated through analysis of the BCL-2 family of proteins. We found a significant increase in the tumor-initiating cell population and increased expression of the pro-survival family member Bcl-2, which is widely known for its oncogenic behavior. ABT-199 (Venetoclax), is a BH3 mimetic that specifically targets Bcl-2. ABT-199 has been used as a single or combination therapy in multiple hematologic malignancies and has shown promise in multiple subtypes of breast cancer. To address the hypothesis that APC-induced Bcl-2 increase is responsible for PTX resistance, we combined treatment of PTX and ABT-199. This combination treatment of CRISPR-mediated APC knockout MDA-MB-231 cells resulted in alterations in apoptosis, suggesting that Bcl-2 inhibition restores PTX sensitivity in APC knockout breast cancer cells. Our studies are the first to show that Bcl-2 functional inhibition restores PTX sensitivity in APC mutant breast cancer cells. These studies are critical to advance better treatment regimens in patients with TNBC.


Subject(s)
Apoptosis , Drug Resistance, Neoplasm , Proto-Oncogene Proteins c-bcl-2 , Triple Negative Breast Neoplasms , Humans , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Apoptosis/drug effects , Female , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Sulfonamides/pharmacology , Paclitaxel/pharmacology , Up-Regulation/drug effects , Taxoids/pharmacology , Bridged-Ring Compounds , Bridged Bicyclo Compounds, Heterocyclic
10.
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
11.
Genome Med ; 16(1): 99, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138508

ABSTRACT

BACKGROUND: There are known disparities in incidence and outcomes of colorectal cancer (CRC) by race and ethnicity. Some of these disparities may be mediated by molecular changes in tumors that occur at different rates across populations. Genetic ancestry is a measure complementary to race and ethnicity that can overcome missing data issues and better capture genetic similarity in admixed populations. We aimed to identify somatic mutations and tumor gene expression differences associated with both genetic ancestry and imputed race and ethnicity. METHODS: Sequencing was performed with the Tempus xT NGS 648-gene panel and whole exome capture RNA-Seq for 8454 primarily late-stage CRC patients. Genetic ancestry proportions for five continental groups-Africa (AFR), American indigenous (AMR), East Asia (EAS), Europe (EUR), and South Asia (SAS)-were estimated using ancestry informative markers. To address data gaps, race and ethnicity categories were imputed, resulting in assignments for 952 Hispanic/Latino, 420 non-Hispanic (NH) Asian, 1061 NH Black, and 5763 NH White individuals. We assessed association of genetic ancestry proportions and imputed race and ethnicity categories with somatic mutations in relevant CRC genes and in 2608 expression profiles, as well as 1957 consensus molecular subtypes (CMS). RESULTS: Increased AFR ancestry was associated with higher odds of somatic mutations in APC, KRAS, and PIK3CA and lower odds of BRAF mutations. Additionally, increased EAS ancestry was associated with lower odds of mutations in KRAS, EUR with higher odds in BRAF, and the Hispanic/Latino category with lower odds in BRAF. Greater AFR ancestry and the NH Black category were associated with higher rates of CMS3, while a higher proportion of Hispanic/Latino patients exhibited indeterminate CMS classifications. CONCLUSIONS: Molecular differences in CRC tumor mutation frequencies and gene expression that may underlie observed differences by race and ethnicity were identified. The association of AFR ancestry with increased KRAS mutations aligns with higher CMS3 subtype rates in NH Black patients. The increase of indeterminate CMS in Hispanic/Latino patients suggests that subtype classification methods could benefit from enhanced patient diversity.


Subject(s)
Colorectal Neoplasms , Mutation , Humans , Colorectal Neoplasms/genetics , Male , Female , Middle Aged , Proto-Oncogene Proteins B-raf/genetics , Aged , Class I Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Biomarkers, Tumor/genetics , Adenomatous Polyposis Coli Protein/genetics
12.
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
13.
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
14.
Eur Urol Oncol ; 7(3): 597-604, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38182487

ABSTRACT

BACKGROUND: Visceral metastases are known to occur in advanced prostate cancer, usually when the tumour is resistant to androgen deprivation and, have worse outcomes regardless of therapies. OBJECTIVE: To analyse genomic alterations in tumour samples according to their lymphatic, bone, and visceral metastatic stages and overall survival. DESIGN, SETTING, AND PARTICIPANTS: We selected 200 patients with metastatic prostate cancer. Genomic profiling of 111 genes and molecular signatures (homologous recombination deficiency [HRD], microsatellite instability, and tumour burden mutation) was performed with the MyChoice test (Myriad Genetics, Inc, Salt Lake City, UT, USA). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: The association between genomic profiles and visceral metastatic evolution was evaluated using logistic regression. Kaplan-Meier and Cox proportional hazard analyses were used for analyses of early death. RESULTS AND LIMITATIONS: A total of 173 (87%) genomic profiles were obtained. Eighty-four (49%) patients died during the follow-up period (median duration = 76 mo). TP53 was the most frequently mutated gene, followed by FANC genes, including BRCA2, and those of the Wnt-pathway (APC/CTNNB1). TP53 gene mutations were more frequent in patients of European (42%) than in those of African (16%) ancestry. An HRD score of >25 was predictive of FANC gene mutations. The mutational status of TP53 (p < 0.001) and APC (p = 0.002) genes were significantly associated with the risk of visceral metastases. The mutational status of CTNNB1 (p = 0.001), TP53 (p = 0.015), BRCA2 (p = 0.027), and FANC (p = 0.005) genes were significantly associated with an earlier age at death. The limitations are the retrospective study design based on a selection of genes and the low frequency of certain molecular events. CONCLUSIONS: Mutations in the TP53 gene and genes (APC/CTNNB1) related to the Wnt pathway are associated with metastatic visceral dissemination and early death. These genomic alterations could be considered as markers to identify prostate cancer patients at a high risk of life-threatening disease who might benefit from more intensified treatment or new targeted therapies. PATIENT SUMMARY: In this report, we evaluated the relationships between genomic profiles (gene mutations and molecular signatures) of tumour samples from patients with metastatic prostate cancer and early death. We found that mutations of specific genes, notably TP53 and APC/CTNNB1 related to the Wnt pathway, are associated with visceral metastatic progression and an earlier age at death.


Subject(s)
Mutation , Prostatic Neoplasms , Humans , Male , Aged , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/mortality , Middle Aged , Aged, 80 and over , Adenomatous Polyposis Coli Protein/genetics , beta Catenin/genetics , Tumor Suppressor Protein p53/genetics , Prognosis , BRCA2 Protein/genetics , Kaplan-Meier Estimate , Bone Neoplasms/secondary , Bone Neoplasms/genetics , Bone Neoplasms/mortality , Neoplasm Metastasis , Proportional Hazards Models , Lymphatic Metastasis/genetics , Microsatellite Instability
15.
Lung Cancer ; 192: 107825, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38795461

ABSTRACT

Pulmonary large cell neuroendocrine carcinoma (LCNEC) is a highly aggressive neoplasm with biological heterogeneity. Mutations in multiple genes have been identified in LCNEC. However, associations between gene alterations, histopathological characteristics, and prognosis remain ambiguous. Here, we investigated the clinicopathologic, immunohistochemical, and genomic characteristics of 19 patients with LCNEC and 9 patients with atypical carcinoid (AC). We revealed high mutation frequencies of TP53 (89.5 %), RB1 (42.1 %), APC (31.6 %), and MCL1 (31.6 %) in LCNEC, while genetic alterations were rarely found in AC. APC alterations mainly occurred to the exon 16 and were only identified in LCNEC with wild-type RB1. The 19 LCNEC were further subgrouped into APC wild-type (LCNEC-APCMT, 6/19) and APC-mutated (LCNEC-APCWT, 13/19) subgroups. In comparison with LCNEC-APCWT, LCNEC-APCMT displayed lower TMB (median: 12.64 vs 4.20, P = 0.045), and relatively mild cytologic atypia. In addition, LCNEC-APCMT distinguished itself from AC and LCNEC-APCWT by obviously downregulated expression of neuroendocrine markers (CD56 and Syn, P < 0.01) and significantly altered expression of genes downstream of APC (ß-catenin migrating into the cytoplasm and nucleus, P < 0.001; c-Myc upregulating, P = 0.005). The OS of LCNEC-APCMT was numerically intermediate between AC and LCNEC-APCWT. We first proposed that APC alterations were common in LCNEC with wild-type RB1 and that LCNEC-APCMT was associated with lower TMB and better OS in comparison with LCNEC-APCWT.


Subject(s)
Carcinoma, Large Cell , Carcinoma, Neuroendocrine , Lung Neoplasms , Mutation , Humans , Male , Female , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/pathology , Middle Aged , Prognosis , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/mortality , Lung Neoplasms/diagnosis , Aged , Carcinoma, Large Cell/genetics , Carcinoma, Large Cell/pathology , DNA Mutational Analysis , Adult , Biomarkers, Tumor/genetics , Adenomatous Polyposis Coli Protein/genetics , Aged, 80 and over
16.
Methods Mol Biol ; 2828: 57-68, 2024.
Article in English | MEDLINE | ID: mdl-39147970

ABSTRACT

Recent cancer genome analyses have identified frequently mutated genes that are responsible for the development and malignant progression of cancers, including colorectal cancer (CRC). We previously constructed mouse models that carried major driver mutations of CRC, namely Apc, Kras, Tgfbr2, Trp53, and Fbxw7, in combinations. Comprehensive histological analyses of the models showed a link between mutation combinations and malignant phenotypes, such as invasion, epithelial-mesenchymal transition (EMT), and metastasis. The major cause of cancer-related death is metastasis, making it important to understand the mechanism underlying metastasis in order to develop novel therapeutic strategies. To this end, we have established intestinal tumor-derived organoids from different genotyped mice and generated liver metastasis models via transplantation of the organoids into the spleen. Through histological and imaging analyses of the transplantation models, we have determined that the combination of Apc, Kras, Tgfbr2, and Trp53 mutations promotes liver metastasis at a high incidence. We also demonstrated polyclonal metastasis of tumor cell clusters consisting of genetically and phenotypically distinct cells through our model analysis. These organoid transplantation models recapitulate human CRC metastasis, constituting a useful tool for basic and clinical cancer research as a preclinical model. We herein report the experimental protocols of the organoid culture and generation of metastasis models.


Subject(s)
Liver Neoplasms , Mutation , Organoids , Animals , Organoids/pathology , Mice , Liver Neoplasms/secondary , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Genotype , Disease Models, Animal , Tumor Suppressor Protein p53/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Neoplasm Metastasis , Humans , Adenomatous Polyposis Coli Protein/genetics , Receptor, Transforming Growth Factor-beta Type II/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Epithelial-Mesenchymal Transition/genetics
17.
PLoS One ; 19(2): e0295030, 2024.
Article in English | MEDLINE | ID: mdl-38324534

ABSTRACT

Colorectal cancer is the third most common cancer and the second leading cause of cancer-related deaths worldwide. The centrosome is the main microtubule-organizing center in animal cells and centrosome amplification is a hallmark of cancer cells. To investigate the importance of centrosomes in colorectal cancer, we induced centrosome loss in normal and cancer human-derived colorectal organoids using centrinone B, a Polo-like kinase 4 (Plk4) inhibitor. We show that centrosome loss represses human normal colorectal organoid growth in a p53-dependent manner in accordance with previous studies in cell models. However, cancer colorectal organoid lines exhibited different sensitivities to centrosome loss independently of p53. Centrinone-induced cancer organoid growth defect/death positively correlated with a loss of function mutation in the APC gene, suggesting a causal role of the hyperactive WNT pathway. Consistent with this notion, ß-catenin inhibition using XAV939 or ICG-001 partially prevented centrinone-induced death and rescued the growth two APC-mutant organoid lines tested. Our study reveals a novel role for canonical WNT signaling in regulating centrosome loss-induced growth defect/death in a subset of APC-mutant colorectal cancer independently of the classical p53 pathway.


Subject(s)
Adenomatous Polyposis Coli Protein , Colorectal Neoplasms , Tumor Suppressor Protein p53 , beta Catenin , Animals , Humans , beta Catenin/genetics , beta Catenin/metabolism , Centrosome/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Pyrimidines , Sulfones , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Wnt Signaling Pathway , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism
18.
Nat Genet ; 56(7): 1456-1467, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38902475

ABSTRACT

According to conventional views, colon cancer originates from stem cells. However, inflammation, a key risk factor for colon cancer, has been shown to suppress intestinal stemness. Here, we used Paneth cells as a model to assess the capacity of differentiated lineages to trigger tumorigenesis in the context of inflammation in mice. Upon inflammation, Paneth cell-specific Apc mutations led to intestinal tumors reminiscent not only of those arising in patients with inflammatory bowel disease, but also of a larger fraction of human sporadic colon cancers. The latter is possibly because of the inflammatory consequences of western-style dietary habits, a major colon cancer risk factor. Machine learning methods designed to predict the cell-of-origin of cancer from patient-derived tumor samples confirmed that, in a substantial fraction of sporadic cases, the origins of colon cancer reside in secretory lineages and not in stem cells.


Subject(s)
Carcinogenesis , Cell Lineage , Colonic Neoplasms , Inflammation , Paneth Cells , Animals , Mice , Cell Lineage/genetics , Paneth Cells/pathology , Humans , Inflammation/genetics , Inflammation/pathology , Carcinogenesis/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Mutation , Stem Cells/pathology , Cell Differentiation/genetics , Cell Transformation, Neoplastic/genetics , Adenomatous Polyposis Coli Protein/genetics , Mice, Inbred C57BL , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology
19.
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
20.
Toxins (Basel) ; 16(5)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38787064

ABSTRACT

Microcystins (MCs), toxins generated by cyanobacteria, feature microcystin-LR (MC-LR) as one of the most prevalent and toxic variants in aquatic environments. MC-LR not only causes environmental problems but also presents a substantial risk to human health. This study aimed to investigate the impact of MC-LR on APCmin/+ mice, considered as an ideal animal model for intestinal tumors. We administered 40 µg/kg MC-LR to mice by gavage for 8 weeks, followed by histopathological examination, microbial diversity and metabolomics analysis. The mice exposed to MC-LR exhibited a significant promotion in colorectal cancer progression and impaired intestinal barrier function in the APCmin/+ mice compared with the control. Gut microbial dysbiosis was observed in the MC-LR-exposed mice, manifesting a notable alteration in the structure of the gut microbiota. This included the enrichment of Marvinbryantia, Gordonibacter and Family_XIII_AD3011_group and reductions in Faecalibaculum and Lachnoclostridium. Metabolomics analysis revealed increased bile acid (BA) metabolites in the intestinal contents of the mice exposed to MC-LR, particularly taurocholic acid (TCA), alpha-muricholic acid (α-MCA), 3-dehydrocholic acid (3-DHCA), 7-ketodeoxycholic acid (7-KDCA) and 12-ketodeoxycholic acid (12-KDCA). Moreover, we found that Marvinbryantia and Family_XIII_AD3011_group showed the strongest positive correlation with taurocholic acid (TCA) in the mice exposed to MC-LR. These findings provide new insights into the roles and mechanisms of MC-LR in susceptible populations, providing a basis for guiding values of MC-LR in drinking water.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Marine Toxins , Microcystins , Animals , Microcystins/toxicity , Gastrointestinal Microbiome/drug effects , Colorectal Neoplasms/pathology , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Male , Disease Progression , Dysbiosis/chemically induced , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Bile Acids and Salts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL