Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 39
Filtrer
1.
Nat Genet ; 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38902475

RÉSUMÉ

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.

2.
Sci Rep ; 14(1): 11562, 2024 05 21.
Article de Anglais | MEDLINE | ID: mdl-38773237

RÉSUMÉ

Predisposing factors underlying familial aggregation of non-syndromic gliomas are still to be uncovered. Whole-exome sequencing was performed in four Finnish families with brain tumors to identify rare predisposing variants. A total of 417 detected exome variants and 102 previously reported glioma-related variants were further genotyped in 19 Finnish families with brain tumors using targeted sequencing. Rare damaging variants in GALNT13, MYO10 and AR were identified. Two families carried either c.553C>T (R185C) or c.1214T>A (L405Q) on GALNT13. Variant c.553C>T is located on the substrate-binding site of GALNT13. AR c.2180G>T (R727L), which is located on a ligand-binding domain of AR, was detected in two families, one of which also carried a GALNT13 variant. MYO10 c.4448A>G (N1483S) was detected in two families and c.1511C>T (A504V) variant was detected in one family. Both variants are located on functional domains related to MYO10 activity in filopodia formation. In addition, affected cases in six families carried a known glioma risk variant rs55705857 in CCDC26 and low-risk glioma variants. These novel findings indicate polygenic inheritance of familial glioma in Finland and increase our understanding of the genetic contribution to familial glioma susceptibility.


Sujet(s)
Prédisposition génétique à une maladie , Gliome , N-acetylgalactosaminyltransferase , Pedigree , Humains , Finlande , Gliome/génétique , Gliome/anatomopathologie , Femelle , Mâle , N-acetylgalactosaminyltransferase/génétique , , Mutation germinale , Adulte , Adulte d'âge moyen , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie ,
3.
Int J Cancer ; 154(11): 1987-1998, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38319157

RÉSUMÉ

Approximately 5% of colorectal cancers (CRCs) have a gain-of-function mutation in the GNAS gene, which leads to the activation of cAMP-dependent signaling pathways and associates with poor prognosis. We investigated the effect of an activating GNAS mutation in CRC cell lines on gene expression and cell proliferation in vitro, and tumor growth in vivo. GNAS-mutated (GNASmt) HCT116 cells showed stimulated synthesis of cAMP as compared to parental (Par) cells. The most upregulated gene in the GNASmt cells was cAMP-hydrolyzing phosphodiesterase 4D (PDE4D) as detected by RNA sequencing. To further validate our finding, we analyzed PDE4D expression in a set of human CRC tumors (n = 35) and demonstrated overexpression in GNAS mutant CRC tumors as compared to GNAS wild-type tumors. The GNASmt HCT116 cells proliferated more slowly than the Par cells. PDE4 inhibitor Ro 20-1724 and PDE4D subtype selective inhibitor GEBR-7b further suppressed the proliferation of GNASmt cells without an effect on Par cells. The growth inhibitory effect of these inhibitors was also seen in the intrinsically GNAS-mutated SK-CO-1 CRC cell line having high levels of cAMP synthesis and PDE4D expression. In vivo, GNASmt HCT116 cells formed smaller tumors than the Par cells in nude mice. In conclusion, our findings demonstrate that GNAS mutation results in the growth suppression of CRC cells. Moreover, the GNAS mutation-induced overexpression of PDE4D provides a potential avenue to impede the proliferation of CRC cells through the use of PDE4 inhibitors.


Sujet(s)
Chromogranine , Tumeurs colorectales , Cyclic Nucleotide Phosphodiesterases, Type 4 , Sous-unités alpha Gs des protéines G , Animaux , Humains , Souris , Chromogranine/génétique , Chromogranine/métabolisme , Tumeurs colorectales/génétique , Cyclic Nucleotide Phosphodiesterases, Type 4/génétique , Cyclic Nucleotide Phosphodiesterases, Type 4/métabolisme , Sous-unités alpha Gs des protéines G/génétique , Sous-unités alpha Gs des protéines G/métabolisme , Cellules HCT116 , Souris nude , Mutation , Inhibiteurs de la phosphodiestérase-4/pharmacologie
4.
bioRxiv ; 2023 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-37873142

RÉSUMÉ

According to conventional views, colon cancer originates from stem cells. However, inflammation, a key risk factor for colon cancer, was shown to suppress intestinal stemness. Here, we employed Paneth cells (PCs) as a model to assess the capacity of differentiated lineages to trigger tumorigenesis in the context of inflammation. Upon inflammation, PC-specific Apc mutations led to intestinal tumors reminiscent not only of those arising in inflammatory bowel disease (IBD) patients but also of a larger fraction of sporadic colon cancers. The latter is likely due to the inflammatory consequences of Western-style dietary habits, the major colon cancer risk factor. Computational methods designed to predict the cell-of-origin of cancer confirmed that, in a substantial fraction of sporadic colon cancers the cells-of-origin are secretory lineages and not stem cells.

5.
Gastroenterology ; 165(4): 861-873, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37453564

RÉSUMÉ

BACKGROUND & AIMS: Small intestinal neuroendocrine tumor (SI-NET) is a rare disease, but its incidence has increased over the past 4 decades. Understanding the genetic risk factors underlying SI-NETs can help in disease prevention and may provide clinically beneficial markers for diagnosis. Here the results of the largest genome-wide association study of SI-NETs performed to date with 405 cases and 614,666 controls are reported. METHODS: Samples from 307 patients with SI-NETs and 287,137 controls in the FinnGen study were used for the identification of SI-NET risk-associated genetic variants. The results were also meta-analyzed with summary statistics from the UK Biobank (n = 98 patients with SI-NET and n = 327,529 controls). RESULTS: We identified 6 genome-wide significant (P < 5 × 10-8) loci associated with SI-NET risk, of which 4 (near SEMA6A, LGR5, CDKAL1, and FERMT2) are novel and 2 (near LTA4H-ELK and in KIF16B) have been reported previously. Interestingly, the top hit (rs200138614; P = 1.80 × 10-19) was a missense variant (p.Cys712Phe) in the LGR5 gene, a bona-fide marker of adult intestinal stem cells and a potentiator of canonical WNT signaling. The association was validated in an independent Finnish collection of 70 patients with SI-NETs, as well as in the UK Biobank exome sequence data (n = 92 cases and n = 392,814 controls). Overexpression of LGR5 p.Cys712Phe in intestinal organoids abolished the ability of R-Spondin1 to support organoid growth, indicating that the mutation perturbed R-Spondin-LGR5 signaling. CONCLUSIONS: Our study is the largest genome-wide association study to date on SI-NETs and reported 4 new associated genome-wide association study loci, including a novel missense mutation (rs200138614, p.Cys712Phe) in LGR5, a canonical marker of adult intestinal stem cells.


Sujet(s)
Tumeurs de l'intestin , Tumeurs neuroendocrines , Adulte , Humains , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/anatomopathologie , Mutation faux-sens , Étude d'association pangénomique , Tumeurs de l'intestin/génétique , Tumeurs de l'intestin/anatomopathologie , Récepteurs couplés aux protéines G/génétique , Kinésine/génétique
6.
Am J Hum Genet ; 110(3): 460-474, 2023 03 02.
Article de Anglais | MEDLINE | ID: mdl-36773604

RÉSUMÉ

Uterine leiomyomas (ULs) are benign smooth muscle tumors that are common in premenopausal women. Somatic alterations in MED12, HMGA2, FH, genes encoding subunits of the SRCAP complex, and genes involved in Cullin 3-RING E3 ligase neddylation are mutually exclusive UL drivers. Established predisposition genes explain only partially the estimated heritability of leiomyomas. Here, we examined loss-of-function variants across 18,899 genes in a cohort of 233,614 White European women, revealing variants in four genes encoding SRCAP complex subunits (YEATS4, ZNHIT1, DMAP1, and ACTL6A) with a significant association to ULs, and YEATS4 and ZNHIT1 strikingly rank first and second, respectively. Positive mutation status was also associated with younger age at diagnosis and hysterectomy. Moderate-penetrance UL risk was largely attributed to rare non-synonymous mutations affecting the SRCAP complex. To examine this disease phenotype more closely, we set out to identify inherited mutations affecting the SRCAP complex in our in-house sample collection of Finnish individuals with ULs (n = 860). We detected one individual with an ACTL6A splice-site mutation, two individuals with a YEATS4 missense mutation, and four individuals with DMAP1 mutations: one splice-site, one nonsense, and two missense variants. These individuals had large and/or multiple ULs, were often diagnosed at an early age, and many had family history of ULs. When a somatic second hit was found, ACTL6A and DMAP1 were silenced in tumors by somatic mutation and YEATS4 by promoter hypermethylation. Decreased H2A.Z staining was observed in the tumors, providing further evidence for the pathogenic nature of the germline mutations. Our results establish inactivation of genes encoding SRCAP complex subunits as a central contributor to moderate-penetrance UL predisposition.


Sujet(s)
Léiomyome , Tumeurs de l'utérus , Humains , Femelle , Tumeurs de l'utérus/génétique , Tumeurs de l'utérus/anatomopathologie , Mutation germinale , Pénétrance , Analyse de mutations d'ADN , Léiomyome/génétique , Léiomyome/anatomopathologie , Mutation , Complexe médiateur/génétique , Actines/génétique , Protéines chromosomiques nonhistones/génétique , Protéines de liaison à l'ADN/génétique , Adenosine triphosphatases/génétique
7.
Res Sq ; 2023 Jan 19.
Article de Anglais | MEDLINE | ID: mdl-36711533

RÉSUMÉ

Paneth cells (PCs), responsible for the secretion of antimicrobial peptides in the small intestine and for niche support to Lgr5+ crypt-base columnar stem cells (CBCs), have been shown to respond to inflammation by dedifferentiating into stem-like cells in order to sustain a regenerative response1,2. Therefore, PCs may represent the cells-of-origin of intestinal cancer in the context of inflammation. To test this hypothesis, we targeted Apc, Kras, and Tp53 mutations in Paneth cells by Cre-Lox technology and modelled inflammation by dextran sodium sulfate (DSS) administration. PC-specific loss of Apc resulted in multiple small intestinal tumors, whereas Kras or Tp53 mutations did not. Compound Apc and Kras mutations in PCs resulted in a striking increase in tumor multiplicity even in the absence of the inflammatory insult. By combining scRNAseq with lineage tracing to capture the conversion of PCs into bona fide tumor cells, we show that they progress through a "revival stem cell" (RSC) state characterized by high Clusterin (Clu) expression and Yap1 signaling, reminiscent of what has been previously observed upon irradiation of the mouse digestive tract3. Accordingly, comparison of PC- and Lgr5-derived murine intestinal tumors revealed differences related to Wnt signaling and inflammatory pathways which match the dichotomy of CBCs and injury-induced RSCs4 between human sporadic colon cancers and those arising in the context of inflammatory bowel diseases. Last, we show that western-style dietary habits, known to trigger a low-grade inflammation throughout the intestinal tract, underlie the analogous dedifferentiation of Paneth cells and their acquisition of stem-like features. Taken together, our results show that intestinal cancer arises in the context of inflammation through the dedifferentiation of committed secretory lineages such as Paneth cells and the activation of the revival stem cell state. As such, a true quiescent stem cell identity may be hidden in fully committed and postmitotic lineages which, upon inflammation, support the regenerative response by re-entering the cell cycle and dedifferentiating into RSCs. The chronic nature of the tissue insult in inflammatory bowel diseases and even in the context of western-style dietary habits is likely to result in the expansion of cell targets for tumor initiation and progression.

8.
Clin Epigenetics ; 15(1): 7, 2023 01 14.
Article de Anglais | MEDLINE | ID: mdl-36639817

RÉSUMÉ

BACKGROUND: Accurate regulation of DNA methylation is necessary for normal cells to differentiate, develop and function. TET2 catalyzes stepwise DNA demethylation in hematopoietic cells. Mutations in the TET2 gene predispose to hematological malignancies by causing DNA methylation overload and aberrant epigenomic landscape. Studies on mice and cell lines show that the function of TET2 is boosted by vitamin C. Thus, by strengthening the demethylation activity of TET2, vitamin C could play a role in the prevention of hematological malignancies in individuals with TET2 dysfunction. We recently identified a family with lymphoma predisposition where a heterozygous truncating germline mutation in TET2 segregated with nodular lymphocyte-predominant Hodgkin lymphoma. The mutation carriers displayed a hypermethylation pattern that was absent in the family members without the mutation. METHODS: In a clinical trial of 1 year, we investigated the effects of oral 1 g/day vitamin C supplementation on DNA methylation by analyzing genome-wide DNA methylation and gene expression patterns from the family members. RESULTS: We show that vitamin C reinforces the DNA demethylation cascade, reduces the proportion of hypermethylated loci and diminishes gene expression differences between TET2 mutation carriers and control individuals. CONCLUSIONS: These results suggest that vitamin C supplementation increases DNA methylation turnover and provide a basis for further work to examine the potential benefits of vitamin C supplementation in individuals with germline and somatic TET2 mutations. TRIAL REGISTRATION: This trial was registered at EudraCT with reference number of 2018-000155-41 (01.04.2019).


Sujet(s)
Acide ascorbique , Protéines de liaison à l'ADN , Dioxygenases , Tumeurs hématologiques , Acide ascorbique/usage thérapeutique , Dioxygenases/génétique , Déméthylation de l'ADN , Méthylation de l'ADN , Protéines de liaison à l'ADN/génétique , Mutation germinale , Tumeurs hématologiques/génétique , Mutation , Vitamines/usage thérapeutique , Humains
9.
Hum Mol Genet ; 32(7): 1063-1071, 2023 03 20.
Article de Anglais | MEDLINE | ID: mdl-36048862

RÉSUMÉ

Precision medicine carries great potential for management of all tumor types. The aim of this retrospective study was to investigate if the two most common genetically distinct uterine fibroid subclasses, driven by aberrations in MED12 and HMGA2 genes, respectively, influence response to treatment with the progesterone receptor modulator ulipristal acetate. Changes in diameter and mutation status were derived for 101 uterine fibroids surgically removed after ulipristal acetate treatment. A significant difference in treatment response between the two major subclasses was detected. MED12 mutant fibroids had 4.4 times higher odds of shrinking in response to ulipristal acetate treatment as compared to HMGA2 driven fibroids (95% confidence interval 1.37-13.9; P = 0.013), and in a multivariate analysis molecular subclassification was an independent predictive factor. Compatible with this finding, gene expression and DNA methylation analyses revealed subclass specific differences in progesterone receptor signaling. The work provides a proof-of-principle that uterine fibroid treatment response is influenced by molecular subclass and that the genetic subclasses should be taken into account when evaluating current and future uterine fibroid therapies.


Sujet(s)
Léiomyome , Tumeurs de l'utérus , Femelle , Humains , Tumeurs de l'utérus/traitement médicamenteux , Tumeurs de l'utérus/génétique , Tumeurs de l'utérus/anatomopathologie , Récepteurs à la progestérone/génétique , Récepteurs à la progestérone/métabolisme , Études rétrospectives , Léiomyome/traitement médicamenteux , Léiomyome/génétique , Léiomyome/anatomopathologie , Facteurs de transcription
10.
Am J Surg Pathol ; 46(8): 1126-1136, 2022 08 01.
Article de Anglais | MEDLINE | ID: mdl-35426837

RÉSUMÉ

Uterine leiomyoma (UL) is a common benign neoplasm which can sometimes be difficult to differentiate from the uterine inflammatory myofibroblastic tumor (IMT) based on morphology alone. IMT is a myofibroblastic/fibroblastic neoplasm which has typically been considered to be rare in the uterus. Its clinical behavior is usually indolent although aggressive variants exist. The majority of IMTs harbor genomic rearrangement of anaplastic lymphoma kinase ( ALK ), while ALK fusion has not been thus far detected in ULs. We analyzed 2263 ULs of which 9 (0.4%) had tyrosine-kinase activation. Seven of the samples were ALK immunopositive: 6 had an ALK fusion gene and 1 overexpressed an ALK transcript skipping exons 2 to 3, Moreover, 1 sample had a RET , and 1 a PDGFRB fusion gene. While no recurrent somatic mutations were found, 1 patient had an ALK germline mutation. Seven tumors showed leiomyoma-like morphology, 1 tumor had slightly loose, and 1 fibrous growth pattern. Six tumors had mild to moderate lymphocyte infiltration, while no immune cell infiltration was detected in 3 cases. None of the tumors showed aggressive behavior. Except for strong ALK positivity (7/9 tumors) the protein expression profile of the tumors was identical to ULs and distinct from other mesenchymal uterine tumors. In gene expression level, these tumors and the known UL subclasses did not separate perfectly. However, vitamin C metabolism and epithelial-mesenchymal transition pathways were uniquely enriched in these lesions. The overall similarity of the analyzed tumors to UL raises the question whether an UL diagnosis would be more proper for a subset of uterine IMTs.


Sujet(s)
Granulome à plasmocytes , Léiomyome , Tumeurs du tissu conjonctif et des tissus mous , Tumeurs de l'utérus , Femelle , Fusion de gènes , Granulome à plasmocytes/génétique , Humains , Léiomyome/génétique , Protein-tyrosine kinases/génétique , Tumeurs de l'utérus/anatomopathologie
11.
Nat Commun ; 12(1): 5448, 2021 09 14.
Article de Anglais | MEDLINE | ID: mdl-34521855

RÉSUMÉ

Mechanical forces in a constrained cellular environment were recently established as a facilitator of chromosomal damage. Whether this could contribute to tumorigenesis is not known. Uterine leiomyomas are common neoplasms that display relatively few chromosomal aberrations. We hypothesized that if mechanical forces contribute to chromosomal damage, signs of this could be seen in uterine leiomyomas from parous women. We examined the karyotypes of 1946 tumors, and found a striking overrepresentation of chromosomal damage associated with parity. We then subjected myometrial cells to physiological forces similar to those encountered during pregnancy, and found this to cause DNA breaks and a DNA repair response. While mechanical forces acting in constrained cellular environments may thus contribute to neoplastic degeneration, and genesis of uterine leiomyoma, further studies are needed to prove possible causality of the observed association. No evidence for progression to malignancy was found.


Sujet(s)
Aberrations des chromosomes , Réparation de l'ADN , Léiomyome/génétique , Complexe médiateur/génétique , Parité , Tumeurs de l'utérus/génétique , Adulte , Phénomènes biomécaniques , Cassures double-brin de l'ADN , Femelle , Expression des gènes , Humains , Hystérectomie , Caryotype , Léiomyome/étiologie , Léiomyome/anatomopathologie , Léiomyome/chirurgie , Mutation , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Myomètre/métabolisme , Myomètre/anatomopathologie , Grossesse , Culture de cellules primaires , Études prospectives , Tumeurs de l'utérus/étiologie , Tumeurs de l'utérus/anatomopathologie , Tumeurs de l'utérus/chirurgie
12.
Nature ; 596(7872): 398-403, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-34349258

RÉSUMÉ

One in four women suffers from uterine leiomyomas (ULs)-benign tumours of the uterine wall, also known as uterine fibroids-at some point in premenopausal life. ULs can cause excessive bleeding, pain and infertility1, and are a common cause of hysterectomy2. They emerge through at least three distinct genetic drivers: mutations in MED12 or FH, or genomic rearrangement of HMGA23. Here we created genome-wide datasets, using DNA, RNA, assay for transposase-accessible chromatin (ATAC), chromatin immunoprecipitation (ChIP) and HiC chromatin immunoprecipitation (HiChIP) sequencing of primary tissues to profoundly understand the genesis of UL. We identified somatic mutations in genes encoding six members of the SRCAP histone-loading complex4, and found that germline mutations in the SRCAP members YEATS4 and ZNHIT1 predispose women to UL. Tumours bearing these mutations showed defective deposition of the histone variant H2A.Z. In ULs, H2A.Z occupancy correlated positively with chromatin accessibility and gene expression, and negatively with DNA methylation, but these correlations were weak in tumours bearing SRCAP complex mutations. In these tumours, open chromatin emerged at transcription start sites where H2A.Z was lost, which was associated with upregulation of genes. Furthermore, YEATS4 defects were associated with abnormal upregulation of bivalent embryonic stem cell genes, as previously shown in mice5. Our work describes a potential mechanism of tumorigenesis-epigenetic instability caused by deficient H2A.Z deposition-and suggests that ULs arise through an aberrant differentiation program driven by deranged chromatin, emanating from a small number of mutually exclusive driver mutations.


Sujet(s)
Assemblage et désassemblage de la chromatine , Chromatine/génétique , Chromatine/métabolisme , Histone/déficit , Léiomyome/génétique , Mutation , Tumeurs de l'utérus/génétique , Carcinogenèse/génétique , Lignée cellulaire , Chromatine/composition chimique , Cellules souches embryonnaires/métabolisme , Épigenèse génétique , Femelle , Régulation de l'expression des gènes tumoraux , Histone/génétique , Histone/métabolisme , Humains , Léiomyome/métabolisme , Léiomyome/anatomopathologie , Ligases/génétique , Complexe répresseur Polycomb-1/génétique , Protéines du groupe Polycomb/génétique , Facteurs de transcription/génétique , Tumeurs de l'utérus/métabolisme , Tumeurs de l'utérus/anatomopathologie
13.
Gastroenterology ; 161(2): 592-607, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-33930428

RÉSUMÉ

BACKGROUND & AIMS: Inflammatory bowel disease (IBD) is a chronic, relapsing inflammatory disorder associated with an elevated risk of colorectal cancer (CRC). IBD-associated CRC (IBD-CRC) may represent a distinct pathway of tumorigenesis compared to sporadic CRC (sCRC). Our aim was to comprehensively characterize IBD-associated tumorigenesis integrating multiple high-throughput approaches, and to compare the results with in-house data sets from sCRCs. METHODS: Whole-genome sequencing, single nucleotide polymorphism arrays, RNA sequencing, genome-wide methylation analysis, and immunohistochemistry were performed using fresh-frozen and formalin-fixed tissue samples of tumor and corresponding normal tissues from 31 patients with IBD-CRC. RESULTS: Transcriptome-based tumor subtyping revealed the complete absence of canonical epithelial tumor subtype associated with WNT signaling in IBD-CRCs, dominated instead by mesenchymal stroma-rich subtype. Negative WNT regulators AXIN2 and RNF43 were strongly down-regulated in IBD-CRCs and chromosomal gains at HNF4A, a negative regulator of WNT-induced epithelial-mesenchymal transition (EMT), were less frequent compared to sCRCs. Enrichment of hypomethylation at HNF4α binding sites was detected solely in sCRC genomes. PIGR and OSMR involved in mucosal immunity were dysregulated via epigenetic modifications in IBD-CRCs. Genome-wide analysis showed significant enrichment of noncoding mutations to 5'untranslated region of TP53 in IBD-CRCs. As reported previously, somatic mutations in APC and KRAS were less frequent in IBD-CRCs compared to sCRCs. CONCLUSIONS: Distinct mechanisms of WNT pathway dysregulation skew IBD-CRCs toward mesenchymal tumor subtype, which may affect prognosis and treatment options. Increased OSMR signaling may favor the establishment of mesenchymal tumors in patients with IBD.


Sujet(s)
Marqueurs biologiques tumoraux/génétique , Transformation cellulaire néoplasique/génétique , Néoplasmes associés aux colites/génétique , Méthylation de l'ADN , Épigenèse génétique , Maladies inflammatoires intestinales/génétique , Transcriptome , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Transformation cellulaire néoplasique/immunologie , Transformation cellulaire néoplasique/anatomopathologie , Néoplasmes associés aux colites/immunologie , Néoplasmes associés aux colites/anatomopathologie , Analyse de mutations d'ADN , Épigénomique , Femelle , Finlande , Analyse de profil d'expression de gènes , Séquençage nucléotidique à haut débit , Humains , Maladies inflammatoires intestinales/immunologie , Maladies inflammatoires intestinales/anatomopathologie , Mâle , Adulte d'âge moyen , Mutation , Grading des tumeurs , Stadification tumorale , Séquençage par oligonucléotides en batterie , Polymorphisme de nucléotide simple , Analyse de séquence d'ARN , Microenvironnement tumoral/génétique , Microenvironnement tumoral/immunologie , Séquençage du génome entier
14.
Genes Chromosomes Cancer ; 60(7): 463-473, 2021 07.
Article de Anglais | MEDLINE | ID: mdl-33527622

RÉSUMÉ

Microsatellite instability (MSI) is caused by defective DNA mismatch repair (MMR), and manifests as accumulation of small insertions and deletions (indels) in short tandem repeats of the genome. Another form of repeat instability, elevated microsatellite alterations at selected tetranucleotide repeats (EMAST), has been suggested to occur in 50% to 60% of colorectal cancer (CRC), of which approximately one quarter are accounted for by MSI. Unlike for MSI, the criteria for defining EMAST is not consensual. EMAST CRCs have been suggested to form a distinct subset of CRCs that has been linked to a higher tumor stage, chronic inflammation, and poor prognosis. EMAST CRCs not exhibiting MSI have been proposed to show instability of di- and trinucleotide repeats in addition to tetranucleotide repeats, but lack instability of mononucleotide repeats. However, previous studies on EMAST have been based on targeted analysis of small sets of marker repeats, often in relatively few samples. To gain insight into tetranucleotide instability on a genome-wide level, we utilized whole genome sequencing data from 227 microsatellite stable (MSS) CRCs, 18 MSI CRCs, 3 POLE-mutated CRCs, and their corresponding normal samples. As expected, we observed tetranucleotide instability in all MSI CRCs, accompanied by instability of mono-, di-, and trinucleotide repeats. Among MSS CRCs, some tumors displayed more microsatellite mutations than others as a continuum, and no distinct subset of tumors with the previously proposed molecular characters of EMAST could be observed. Our results suggest that tetranucleotide repeat mutations in non-MSI CRCs represent stochastic mutation events rather than define a distinct CRC subclass.


Sujet(s)
Tumeurs colorectales/génétique , Dépistage génétique/méthodes , Mutation de type INDEL , Répétitions microsatellites , Séquençage du génome entier/méthodes , Dépistage génétique/statistiques et données numériques , Humains , Séquençage du génome entier/statistiques et données numériques
16.
Mol Cancer Res ; 17(12): 2432-2443, 2019 12.
Article de Anglais | MEDLINE | ID: mdl-31578227

RÉSUMÉ

Somatic driver mechanisms of pituitary adenoma pathogenesis have remained incompletely characterized; apart from mutations in the stimulatory Gα protein (Gαs encoded by GNAS) causing activated cAMP synthesis, pathogenic variants are rarely found in growth hormone-secreting pituitary tumors (somatotropinomas). The purpose of the current work was to clarify how genetic and epigenetic alterations contribute to the development of somatotropinomas by conducting an integrated copy number alteration, whole-genome and bisulfite sequencing, and transcriptome analysis of 21 tumors. Somatic mutation burden was low, but somatotropinomas formed two subtypes associated with distinct aneuploidy rates and unique transcription profiles. Tumors with recurrent chromosome aneuploidy (CA) were GNAS mutation negative (Gsp- ). The chromosome stable (CS) -group contained Gsp+ somatotropinomas and two totally aneuploidy-free Gsp- tumors. Genes related to the mitotic G1-S-checkpoint transition were differentially expressed in CA- and CS-tumors, indicating difference in mitotic progression. Also, pituitary tumor transforming gene 1 (PTTG1), a regulator of sister chromatid segregation, showed abundant expression in CA-tumors. Moreover, somatotropinomas displayed distinct Gsp genotype-specific methylation profiles and expression quantitative methylation (eQTM) analysis revealed that inhibitory Gα (Gαi) signaling is activated in Gsp+ tumors. These findings suggest that aneuploidy through modulated driver pathways may be a causative mechanism for tumorigenesis in Gsp- somatotropinomas, whereas Gsp+ tumors with constitutively activated cAMP synthesis seem to be characterized by DNA methylation activated Gαi signaling. IMPLICATIONS: These findings provide valuable new information about subtype-specific pituitary tumorigenesis and may help to elucidate the mechanisms of aneuploidy also in other tumor types.


Sujet(s)
Carcinogenèse/génétique , Chromogranine/génétique , Sous-unités alpha Gs des protéines G/génétique , Hormone de croissance/génétique , Tumeurs de l'hypophyse/génétique , Adolescent , Adulte , Sujet âgé , Chromosomes/génétique , AMP cyclique/génétique , Variations de nombre de copies de segment d'ADN/génétique , Méthylation de l'ADN/génétique , Épigenèse génétique , Femelle , Régulation de l'expression des gènes tumoraux/génétique , Génotype , Hormone de croissance/biosynthèse , Humains , Mâle , Adulte d'âge moyen , Mutation , Tumeurs de l'hypophyse/anatomopathologie , Échange de chromatides soeurs/génétique
17.
Nat Commun ; 10(1): 4022, 2019 09 06.
Article de Anglais | MEDLINE | ID: mdl-31492840

RÉSUMÉ

Genomic instability pathways in colorectal cancer (CRC) have been extensively studied, but the role of retrotransposition in colorectal carcinogenesis remains poorly understood. Although retrotransposons are usually repressed, they become active in several human cancers, in particular those of the gastrointestinal tract. Here we characterize retrotransposon insertions in 202 colorectal tumor whole genomes and investigate their associations with molecular and clinical characteristics. We find highly variable retrotransposon activity among tumors and identify recurrent insertions in 15 known cancer genes. In approximately 1% of the cases we identify insertions in APC, likely to be tumor-initiating events. Insertions are positively associated with the CpG island methylator phenotype and the genomic fraction of allelic imbalance. Clinically, high number of insertions is independently associated with poor disease-specific survival.


Sujet(s)
Tumeurs colorectales/génétique , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux , Éléments LINE/génétique , Mutagenèse par insertion , Sujet âgé , Cellules Caco-2 , Carcinogenèse/génétique , Lignée cellulaire tumorale , Tumeurs colorectales/anatomopathologie , Ilots CpG/génétique , Méthylation de l'ADN , Femelle , Instabilité du génome , Humains , Estimation de Kaplan-Meier , Mâle , Adulte d'âge moyen
18.
Clin Infect Dis ; 68(1): 61-69, 2019 01 01.
Article de Anglais | MEDLINE | ID: mdl-29788414

RÉSUMÉ

Background: Different clinical manifestations of invasive pneumococcal disease (IPD) have thus far mainly been explained by patient characteristics. Here we studied the contribution of pneumococcal genetic variation to IPD phenotype. Methods: The index cohort consisted of 349 patients admitted to 2 Dutch hospitals between 2000-2011 with pneumococcal bacteremia. We performed genome-wide association studies to identify pneumococcal lineages, genes, and allelic variants associated with 23 clinical IPD phenotypes. The identified associations were validated in a nationwide (n = 482) and a post-pneumococcal vaccination cohort (n = 121). The contribution of confirmed pneumococcal genotypes to the clinical IPD phenotype, relative to known clinical predictors, was tested by regression analysis. Results: Among IPD patients, the presence of pneumococcal gene slaA was a nationwide confirmed independent predictor of meningitis (odds ratio [OR], 10.5; P = .001), as was sequence cluster 9 (serotype 7F: OR, 3.68; P = .057). A set of 4 pneumococcal genes co-located on a prophage was a confirmed independent predictor of 30-day mortality (OR, 3.4; P = .003). We could detect the pneumococcal variants of concern in these patients' blood samples. Conclusions: In this study, knowledge of pneumococcal genotypic variants improved the clinical risk assessment for detrimental manifestations of IPD. This provides us with novel opportunities to target, anticipate, or avert the pathogenic effects related to particular pneumococcal variants, and indicates that information on pneumococcal genotype is important for the diagnostic and treatment strategy in IPD. Ongoing surveillance is warranted to monitor the clinical value of information on pneumococcal variants in dynamic microbial and susceptible host populations.


Sujet(s)
Bactériémie/microbiologie , Bactériémie/anatomopathologie , Variation génétique , Infections à pneumocoques/microbiologie , Infections à pneumocoques/anatomopathologie , Streptococcus pneumoniae/classification , Streptococcus pneumoniae/génétique , Adolescent , Adulte , Sujet âgé , Études de cohortes , Femelle , Étude d'association pangénomique , Génotype , Humains , Mâle , Adulte d'âge moyen , Appréciation des risques , Sérogroupe , Streptococcus pneumoniae/isolement et purification , Jeune adulte
19.
Elife ; 72018 09 18.
Article de Anglais | MEDLINE | ID: mdl-30226466

RÉSUMÉ

Uterine leiomyomas (ULs) are benign tumors that are a major burden to women's health. A genome-wide association study on 15,453 UL cases and 392,628 controls was performed, followed by replication of the genomic risk in six cohorts. Effects of the risk alleles were evaluated in view of molecular and clinical characteristics. 22 loci displayed a genome-wide significant association. The likely predisposition genes could be grouped to two biological processes. Genes involved in genome stability were represented by TERT, TERC, OBFC1 - highlighting the role of telomere maintenance - TP53 and ATM. Genes involved in genitourinary development, WNT4, WT1, SALL1, MED12, ESR1, GREB1, FOXO1, DMRT1 and uterine stem cell marker antigen CD44, formed another strong subgroup. The combined risk contributed by the 22 loci was associated with MED12 mutation-positive tumors. The findings link genes for uterine development and genetic stability to leiomyomagenesis, and in part explain the more frequent occurrence of UL in women of African origin.


Sujet(s)
Locus génétiques , Prédisposition génétique à une maladie , Instabilité du génome , Léiomyome/génétique , Tumeurs de l'utérus/génétique , Femelle , Étude d'association pangénomique , Humains , Morphogenèse , Appréciation des risques , Utérus/croissance et développement
20.
Nat Commun ; 9(1): 3664, 2018 09 10.
Article de Anglais | MEDLINE | ID: mdl-30202008

RÉSUMÉ

Point mutations in cancer have been extensively studied but chromosomal gains and losses have been more challenging to interpret due to their unspecific nature. Here we examine high-resolution allelic imbalance (AI) landscape in 1699 colorectal cancers, 256 of which have been whole-genome sequenced (WGSed). The imbalances pinpoint 38 genes as plausible AI targets based on previous knowledge. Unbiased CRISPR-Cas9 knockout and activation screens identified in total 79 genes within AI peaks regulating cell growth. Genetic and functional data implicate loss of TP53 as a sufficient driver of AI. The WGS highlights an influence of copy number aberrations on the rate of detected somatic point mutations. Importantly, the data reveal several associations between AI target genes, suggesting a role for a network of lineage-determining transcription factors in colorectal tumorigenesis. Overall, the results unravel the contribution of AI in colorectal cancer and provide a plausible explanation why so few genes are commonly affected by point mutations in cancers.


Sujet(s)
Déséquilibre allélique , Tumeurs colorectales/génétique , Prédisposition génétique à une maladie , Systèmes CRISPR-Cas , Aberrations des chromosomes , Chromosomes humains de la paire 8 , Tumeurs colorectales/anatomopathologie , Variations de nombre de copies de segment d'ADN , Danemark , Analyse de profil d'expression de gènes , Génomique , Génotype , Humains , Perte d'hétérozygotie , Répétitions microsatellites , Phénotype , Mutation ponctuelle , Protéines proto-oncogènes p21(ras)/génétique , Petit ARN interférent/génétique , Facteurs de transcription/génétique , Protéine p53 suppresseur de tumeur/génétique , Séquençage du génome entier
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...