Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 41
Filtrer
1.
HGG Adv ; 5(2): 100273, 2024 Apr 11.
Article de Anglais | MEDLINE | ID: mdl-38297832

RÉSUMÉ

Heterozygous missense variants and in-frame indels in SMC3 are a cause of Cornelia de Lange syndrome (CdLS), marked by intellectual disability, growth deficiency, and dysmorphism, via an apparent dominant-negative mechanism. However, the spectrum of manifestations associated with SMC3 loss-of-function variants has not been reported, leading to hypotheses of alternative phenotypes or even developmental lethality. We used matchmaking servers, patient registries, and other resources to identify individuals with heterozygous, predicted loss-of-function (pLoF) variants in SMC3, and analyzed population databases to characterize mutational intolerance in this gene. Here, we show that SMC3 behaves as an archetypal haploinsufficient gene: it is highly constrained against pLoF variants, strongly depleted for missense variants, and pLoF variants are associated with a range of developmental phenotypes. Among 14 individuals with SMC3 pLoF variants, phenotypes were variable but coalesced on low growth parameters, developmental delay/intellectual disability, and dysmorphism, reminiscent of atypical CdLS. Comparisons to individuals with SMC3 missense/in-frame indel variants demonstrated an overall milder presentation in pLoF carriers. Furthermore, several individuals harboring pLoF variants in SMC3 were nonpenetrant for growth, developmental, and/or dysmorphic features, and some had alternative symptomatologies with rational biological links to SMC3. Analyses of tumor and model system transcriptomic data and epigenetic data in a subset of cases suggest that SMC3 pLoF variants reduce SMC3 expression but do not strongly support clustering with functional genomic signatures of typical CdLS. Our finding of substantial population-scale LoF intolerance in concert with variable growth and developmental features in subjects with SMC3 pLoF variants expands the scope of cohesinopathies, informs on their allelic architecture, and suggests the existence of additional clearly LoF-constrained genes whose disease links will be confirmed only by multilayered genomic data paired with careful phenotyping.


Sujet(s)
Syndrome de Cornelia de Lange , Déficience intellectuelle , Humains , Protéines du cycle cellulaire/génétique , Protéoglycanes à chondroïtine sulfate/génétique , Protéines chromosomiques nonhistones/génétique , Syndrome de Cornelia de Lange/génétique , Hétérozygote , Déficience intellectuelle/génétique , Mutation , Phénotype
3.
medRxiv ; 2023 Sep 28.
Article de Anglais | MEDLINE | ID: mdl-37808847

RÉSUMÉ

Heterozygous missense variants and in-frame indels in SMC3 are a cause of Cornelia de Lange syndrome (CdLS), marked by intellectual disability, growth deficiency, and dysmorphism, via an apparent dominant-negative mechanism. However, the spectrum of manifestations associated with SMC3 loss-of-function variants has not been reported, leading to hypotheses of alternative phenotypes or even developmental lethality. We used matchmaking servers, patient registries, and other resources to identify individuals with heterozygous, predicted loss-of-function (pLoF) variants in SMC3, and analyzed population databases to characterize mutational intolerance in this gene. Here, we show that SMC3 behaves as an archetypal haploinsufficient gene: it is highly constrained against pLoF variants, strongly depleted for missense variants, and pLoF variants are associated with a range of developmental phenotypes. Among 13 individuals with SMC3 pLoF variants, phenotypes were variable but coalesced on low growth parameters, developmental delay/intellectual disability, and dysmorphism reminiscent of atypical CdLS. Comparisons to individuals with SMC3 missense/in-frame indel variants demonstrated a milder presentation in pLoF carriers. Furthermore, several individuals harboring pLoF variants in SMC3 were nonpenetrant for growth, developmental, and/or dysmorphic features, some instead having intriguing symptomatologies with rational biological links to SMC3 including bone marrow failure, acute myeloid leukemia, and Coats retinal vasculopathy. Analyses of transcriptomic and epigenetic data suggest that SMC3 pLoF variants reduce SMC3 expression but do not result in a blood DNA methylation signature clustering with that of CdLS, and that the global transcriptional signature of SMC3 loss is model-dependent. Our finding of substantial population-scale LoF intolerance in concert with variable penetrance in subjects with SMC3 pLoF variants expands the scope of cohesinopathies, informs on their allelic architecture, and suggests the existence of additional clearly LoF-constrained genes whose disease links will be confirmed only by multi-layered genomic data paired with careful phenotyping.

4.
Life Sci Alliance ; 6(10)2023 10.
Article de Anglais | MEDLINE | ID: mdl-37487640

RÉSUMÉ

Polycomb repressive complex 1 (PRC1) strongly influences 3D genome organization, mediating local chromatin compaction and clustering of target loci. Several PRC1 subunits have the capacity to form biomolecular condensates through liquid-liquid phase separation in vitro and when tagged and over-expressed in cells. Here, we use 1,6-hexanediol, which can disrupt liquid-like condensates, to examine the role of endogenous PRC1 biomolecular condensates on local and chromosome-wide clustering of PRC1-bound loci. Using imaging and chromatin immunoprecipitation, we show that PRC1-mediated chromatin compaction and clustering of targeted genomic loci-at different length scales-can be reversibly disrupted by the addition and subsequent removal of 1,6-hexanediol to mouse embryonic stem cells. Decompaction and dispersal of polycomb domains and clusters cannot be solely attributable to reduced PRC1 occupancy detected by chromatin immunoprecipitation following 1,6-hexanediol treatment as the addition of 2,5-hexanediol has similar effects on binding despite this alcohol not perturbing PRC1-mediated 3D clustering, at least at the sub-megabase and megabase scales. These results suggest that weak hydrophobic interactions between PRC1 molecules may have a role in polycomb-mediated genome organization.


Sujet(s)
Chromatine , Protéines de Drosophila , Animaux , Souris , Complexe répresseur Polycomb-1 , Noyau de la cellule , Protéines du groupe Polycomb
5.
Elife ; 122023 02 15.
Article de Anglais | MEDLINE | ID: mdl-36790165

RÉSUMÉ

Centrosomes are orbited by centriolar satellites, dynamic multiprotein assemblies nucleated by Pericentriolar material 1 (PCM1). To study the requirement for centriolar satellites, we generated mice lacking PCM1, a crucial component of satellites. Pcm1-/- mice display partially penetrant perinatal lethality with survivors exhibiting hydrocephalus, oligospermia, and cerebellar hypoplasia, and variably expressive phenotypes such as hydronephrosis. As many of these phenotypes have been observed in human ciliopathies and satellites are implicated in cilia biology, we investigated whether cilia were affected. PCM1 was dispensable for ciliogenesis in many cell types, whereas Pcm1-/- multiciliated ependymal cells and human PCM1-/- retinal pigmented epithelial 1 (RPE1) cells showed reduced ciliogenesis. PCM1-/- RPE1 cells displayed reduced docking of the mother centriole to the ciliary vesicle and removal of CP110 and CEP97 from the distal mother centriole, indicating compromised early ciliogenesis. Similarly, Pcm1-/- ependymal cells exhibited reduced removal of CP110 from basal bodies in vivo. We propose that PCM1 and centriolar satellites facilitate efficient trafficking of proteins to and from centrioles, including the departure of CP110 and CEP97 to initiate ciliogenesis, and that the threshold to trigger ciliogenesis differs between cell types.


Sujet(s)
Centrioles , Cils vibratiles , Animaux , Femelle , Humains , Souris , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Centrioles/métabolisme , Centrosome/métabolisme , Cils vibratiles/métabolisme , Protéines du cytosquelette/métabolisme
6.
Cell Rep ; 41(6): 111596, 2022 11 08.
Article de Anglais | MEDLINE | ID: mdl-36351380

RÉSUMÉ

Targeting early-stage lung cancer is vital to improve survival. However, the mechanisms and components of the early tumor suppressor response in lung cancer are not well understood. In this report, we study the role of Toll-like receptor 2 (TLR2), a regulator of oncogene-induced senescence, which is a key tumor suppressor response in premalignancy. Using human lung cancer samples and genetically engineered mouse models, we show that TLR2 is active early in lung tumorigenesis, where it correlates with improved survival and clinical regression. Mechanistically, TLR2 impairs early lung cancer progression via activation of cell intrinsic cell cycle arrest pathways and the proinflammatory senescence-associated secretory phenotype (SASP). The SASP regulates non-cell autonomous anti-tumor responses, such as immune surveillance of premalignant cells, and we observe impaired myeloid cell recruitment to lung tumors after Tlr2 loss. Last, we show that administration of a TLR2 agonist reduces lung tumor growth, highlighting TLR2 as a possible therapeutic target.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Souris , Animaux , Humains , Récepteur de type Toll-2/génétique , Récepteur de type Toll-2/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Tumeurs du poumon/génétique , Gènes suppresseurs de tumeur , Poumon/métabolisme , Vieillissement de la cellule/génétique
7.
PLoS One ; 17(11): e0268149, 2022.
Article de Anglais | MEDLINE | ID: mdl-36413568

RÉSUMÉ

Classical aniridia is a congenital and progressive panocular disorder almost exclusively caused by heterozygous loss-of-function variants at the PAX6 locus. We report nine individuals from five families with severe aniridia and/or microphthalmia (with no detectable PAX6 mutation) with ultrarare monoallelic missense variants altering the Arg51 codon of MAB21L1. These mutations occurred de novo in 3/5 families, with the remaining families being compatible with autosomal dominant inheritance. Mice engineered to carry the p.Arg51Leu change showed a highly-penetrant optic disc anomaly in heterozygous animals with severe microphthalmia in homozygotes. Substitutions of the same codon (Arg51) in MAB21L2, a close homolog of MAB21L1, cause severe ocular and skeletal malformations in humans and mice. The predicted nucleotidyltransferase function of MAB21L1 could not be demonstrated using purified protein with a variety of nucleotide substrates and oligonucleotide activators. Induced expression of GFP-tagged wildtype and mutant MAB21L1 in human cells caused only modest transcriptional changes. Mass spectrometry of immunoprecipitated protein revealed that both mutant and wildtype MAB21L1 associate with transcription factors that are known regulators of PAX6 (MEIS1, MEIS2 and PBX1) and with poly(A) RNA binding proteins. Arg51 substitutions reduce the association of wild-type MAB21L1 with TBL1XR1, a component of the NCoR complex. We found limited evidence for mutation-specific interactions with MSI2/Musashi-2, an RNA-binding proteins with effects on many different developmental pathways. Given that biallelic loss-of-function variants in MAB21L1 result in a milder eye phenotype we suggest that Arg51-altering monoallelic variants most plausibly perturb eye development via a gain-of-function mechanism.


Sujet(s)
Aniridie , Microphtalmie , Humains , Animaux , Souris , Microphtalmie/génétique , Facteur de transcription PAX6/génétique , Aniridie/génétique , Mutation faux-sens , Hétérozygote , Facteurs de transcription/génétique , Protéines à homéodomaine/génétique , Protéines de liaison à l'ARN/génétique , Protéines de l'oeil/génétique , Protéines et peptides de signalisation intracellulaire/génétique
8.
Genes (Basel) ; 13(10)2022 10 05.
Article de Anglais | MEDLINE | ID: mdl-36292683

RÉSUMÉ

Anophthalmia (missing eye) describes a failure of early embryonic ocular development. Mutations in a relatively small set of genes account for 75% of bilateral anophthalmia cases, yet 25% of families currently are left without a molecular diagnosis. Here, we report our experimental work that aimed to uncover the developmental and genetic basis of the anophthalmia characterising the X-linked Ie (eye-ear reduction) X-ray-induced allele in mouse that was first identified in 1947. Histological analysis of the embryonic phenotype showed failure of normal eye development after the optic vesicle stage with particularly severe malformation of the ventral retina. Linkage analysis mapped this mutation to a ~6 Mb region on the X chromosome. Short- and long-read whole-genome sequencing (WGS) of affected and unaffected male littermates confirmed the Ie linkage but identified no plausible causative variants or structural rearrangements. These analyses did reduce the critical candidate interval and revealed evidence of multiple variants within the ancestral DNA, although none were found that altered coding sequences or that were unique to Ie. To investigate early embryonic events at a genetic level, we then generated mouse ES cells derived from male Ie embryos and wild type littermates. RNA-seq and accessible chromatin sequencing (ATAC-seq) data generated from cultured optic vesicle organoids did not reveal any large differences in gene expression or accessibility of putative cis-regulatory elements between Ie and wild type. However, an unbiased TF-footprinting analysis of accessible chromatin regions did provide evidence of a genome-wide reduction in binding of transcription factors associated with ventral eye development in Ie, and evidence of an increase in binding of the Zic-family of transcription factors, including Zic3, which is located within the Ie-refined critical interval. We conclude that the refined Ie critical region at chrX: 56,145,000-58,385,000 contains multiple genetic variants that may be linked to altered cis regulation but does not contain a convincing causative mutation. Changes in the binding of key transcription factors to chromatin causing altered gene expression during development, possibly through a subtle mis-regulation of Zic3, presents a plausible cause for the anophthalmia phenotype observed in Ie, but further work is required to determine the precise causative allele and its genetic mechanism.


Sujet(s)
Anophtalmie , Souris , Mâle , Animaux , Anophtalmie/génétique , Séquences d'acides nucléiques régulatrices , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Chromatine , ADN , Protéines à homéodomaine/génétique
9.
Nat Commun ; 13(1): 5609, 2022 09 24.
Article de Anglais | MEDLINE | ID: mdl-36153345

RÉSUMÉ

Human centromeres appear as constrictions on mitotic chromosomes and form a platform for kinetochore assembly in mitosis. Biophysical experiments led to a suggestion that repetitive DNA at centromeric regions form a compact scaffold necessary for function, but this was revised when neocentromeres were discovered on non-repetitive DNA. To test whether centromeres have a special chromatin structure we have analysed the architecture of a neocentromere. Centromere repositioning is accompanied by RNA polymerase II recruitment and active transcription to form a decompacted, negatively supercoiled domain enriched in 'open' chromatin fibres. In contrast, centromerisation causes a spreading of repressive epigenetic marks to surrounding regions, delimited by H3K27me3 polycomb boundaries and divergent genes. This flanking domain is transcriptionally silent and partially remodelled to form 'compact' chromatin, similar to satellite-containing DNA sequences, and exhibits genomic instability. We suggest transcription disrupts chromatin to provide a foundation for kinetochore formation whilst compact pericentromeric heterochromatin generates mechanical rigidity.


Sujet(s)
Hétérochromatine , Histone , Centromère/génétique , Chromatine/génétique , ADN/génétique , ADN satellite , Hétérochromatine/génétique , Histone/génétique , Humains , RNA polymerase II/génétique
10.
Nat Commun ; 13(1): 3053, 2022 06 01.
Article de Anglais | MEDLINE | ID: mdl-35650196

RÉSUMÉ

In addition to central functions in cell adhesion signalling, integrin-associated proteins have wider roles at sites distal to adhesion receptors. In experimentally defined adhesomes, we noticed that there is clear enrichment of proteins that localise to the nucleus, and conversely, we now report that nuclear proteomes contain a class of adhesome components that localise to the nucleus. We here define a nucleo-adhesome, providing experimental evidence for a remarkable scale of nuclear localisation of adhesion proteins, establishing a framework for interrogating nuclear adhesion protein functions. Adding to nuclear FAK's known roles in regulating transcription, we now show that nuclear FAK regulates expression of many adhesion-related proteins that localise to the nucleus and that nuclear FAK binds to the adhesome component and nuclear protein Hic-5. FAK and Hic-5 work together in the nucleus, co-regulating a subset of genes transcriptionally. We demonstrate the principle that there are subcomplexes of nuclear adhesion proteins that cooperate to control transcription.


Sujet(s)
Noyau de la cellule , Protéome , Adhérence cellulaire , Noyau de la cellule/métabolisme , Protéome/métabolisme , Transduction du signal
11.
Cancer Res ; 82(8): 1548-1559, 2022 04 15.
Article de Anglais | MEDLINE | ID: mdl-35074757

RÉSUMÉ

Intrahepatic cholangiocarcinoma (ICC) is an aggressive malignancy of the bile ducts within the liver characterized by high levels of genetic heterogeneity. In the context of such genetic variability, determining which oncogenic mutations drive ICC growth has been difficult, and developing modes of patient stratification and targeted therapies remains challenging. Here we model the interactions between rare mutations with more common driver genes and combine in silico analysis of patient data with highly multiplexed in vivo CRISPR-spCas9 screens to perform a functional in vivo study into the role genetic heterogeneity plays in driving ICC. Novel tumor suppressors were uncovered, which, when lost, cooperate with the RAS oncoprotein to drive ICC growth. Focusing on a set of driver mutations that interact with KRAS to initiate aggressive, sarcomatoid-type ICC revealed that tumor growth relies on Wnt and PI3K signaling. Pharmacologic coinhibition of Wnt and PI3K in vivo impeded ICC growth regardless of mutational profile. Therefore, Wnt and PI3K activity should be considered as a signature by which patients can be stratified for treatment independent of tumor genotype, and inhibitors of these pathways should be levied to treat ICC. SIGNIFICANCE: This work shows that, despite significant genetic heterogeneity, intrahepatic cholangiocarcinoma relies on a limited number of signaling pathways to grow, suggesting common therapeutic vulnerabilities across patients.


Sujet(s)
Tumeurs des canaux biliaires , Cholangiocarcinome , Tumeurs des canaux biliaires/génétique , Tumeurs des canaux biliaires/anatomopathologie , Conduits biliaires intrahépatiques/anatomopathologie , Cholangiocarcinome/génétique , Cholangiocarcinome/anatomopathologie , Hétérogénéité génétique , Humains , Phosphatidylinositol 3-kinases/génétique
12.
FASEB J ; 36(1): e22082, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34918389

RÉSUMÉ

Vitamin D deficiency is associated with risk of several common cancers, including colorectal cancer (CRC). Here we have utilized patient derived epithelial organoids (ex vivo) and CRC cell lines (in vitro) to show that calcitriol (1,25OHD) increased the expression of the CRC tumor suppressor gene, CDH1, at both the transcript and protein level. Whole genome expression analysis demonstrated significant differential expression of a further six genes after 1,25OHD treatment, including genes with established links to carcinogenesis GADD45, EFTUD1 and KIAA1199. Furthermore, gene ontologies relevant to carcinogenesis were enriched by 1,25OHD treatment (e.g., 'regulation of Wnt signaling pathway', 'regulation of cell death'), with common enriched processes across in vitro and ex vivo cultures including 'negative regulation of cell proliferation', 'regulation of cell migration' and 'regulation of cell differentiation'. Our results identify genes and pathways that are modifiable by calcitriol that have links to CRC tumorigenesis. Hence the findings provide potential mechanism to the epidemiological and clinical trial data indicating a causal association between vitamin D and CRC. We suggest there is strong rationale for further well-designed trials of vitamin D supplementation as a novel CRC chemopreventive and chemotherapeutic agent.


Sujet(s)
Antinéoplasiques/pharmacologie , Carcinogenèse/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Protéines tumorales/biosynthèse , Tumeurs/métabolisme , Transcriptome/effets des médicaments et des substances chimiques , Vitamine D/analogues et dérivés , Cellules Caco-2 , Cellules HCT116 , Humains , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Vitamine D/pharmacologie
13.
Science ; 374(6567): eabj3624, 2021 Oct 29.
Article de Anglais | MEDLINE | ID: mdl-34581622

RÉSUMÉ

Inherited genetic factors can influence the severity of COVID-19, but the molecular explanation underpinning a genetic association is often unclear. Intracellular antiviral defenses can inhibit the replication of viruses and reduce disease severity. To better understand the antiviral defenses relevant to COVID-19, we used interferon-stimulated gene (ISG) expression screening to reveal that 2'-5'-oligoadenylate synthetase 1 (OAS1), through ribonuclease L, potently inhibits severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We show that a common splice-acceptor single-nucleotide polymorphism (Rs10774671) governs whether patients express prenylated OAS1 isoforms that are membrane-associated and sense-specific regions of SARS-CoV-2 RNAs or if they only express cytosolic, nonprenylated OAS1 that does not efficiently detect SARS-CoV-2. In hospitalized patients, expression of prenylated OAS1 was associated with protection from severe COVID-19, suggesting that this antiviral defense is a major component of a protective antiviral response.


Sujet(s)
2',5'-Oligoadenylate synthetase/génétique , 2',5'-Oligoadenylate synthetase/métabolisme , COVID-19/génétique , COVID-19/physiopathologie , ARN double brin/métabolisme , ARN viral/métabolisme , SARS-CoV-2/physiologie , Régions 5' non traduites , Cellules A549 , Animaux , COVID-19/enzymologie , COVID-19/immunologie , Chiroptera/génétique , Chiroptera/virologie , Coronaviridae/enzymologie , Coronaviridae/génétique , Coronaviridae/physiologie , Endoribonucleases/métabolisme , Humains , Interférons/immunologie , Isoenzymes/génétique , Isoenzymes/métabolisme , Phosphodiesterases/génétique , Phosphodiesterases/métabolisme , Polymorphisme de nucléotide simple , Prénylation des protéines , ARN double brin/composition chimique , ARN double brin/génétique , ARN viral/composition chimique , ARN viral/génétique , Rétroéléments , SARS-CoV-2/génétique , Indice de gravité de la maladie , Réplication virale
14.
PLoS One ; 16(8): e0256181, 2021.
Article de Anglais | MEDLINE | ID: mdl-34388204

RÉSUMÉ

Identifying causative variants in cis-regulatory elements (CRE) in neurodevelopmental disorders has proven challenging. We have used in vivo functional analyses to categorize rigorously filtered CRE variants in a clinical cohort that is plausibly enriched for causative CRE mutations: 48 unrelated males with a family history consistent with X-linked intellectual disability (XLID) in whom no detectable cause could be identified in the coding regions of the X chromosome (chrX). Targeted sequencing of all chrX CRE identified six rare variants in five affected individuals that altered conserved bases in CRE targeting known XLID genes and segregated appropriately in families. Two of these variants, FMR1CRE and TENM1CRE, showed consistent site- and stage-specific differences of enhancer function in the developing zebrafish brain using dual-color fluorescent reporter assay. Mouse models were created for both variants. In male mice Fmr1CRE induced alterations in neurodevelopmental Fmr1 expression, olfactory behavior and neurophysiological indicators of FMRP function. The absence of another likely causative variant on whole genome sequencing further supported FMR1CRE as the likely basis of the XLID in this family. Tenm1CRE mice showed no phenotypic anomalies. Following the release of gnomAD 2.1, reanalysis showed that TENM1CRE exceeded the maximum plausible population frequency of a XLID causative allele. Assigning causative status to any ultra-rare CRE variant remains problematic and requires disease-relevant in vivo functional data from multiple sources. The sequential and bespoke nature of such analyses renders them time-consuming and challenging to scale for routine clinical use.


Sujet(s)
Protéine du syndrome X fragile/génétique , Gènes liés au chromosome X , Génome humain , Retard mental lié à l'X/génétique , Protéines de tissu nerveux/génétique , Éléments de régulation transcriptionnelle , Ténascine/génétique , Animaux , Animal génétiquement modifié , Encéphale/métabolisme , Encéphale/anatomopathologie , Cartographie chromosomique , Études de cohortes , Modèles animaux de maladie humaine , Embryon non mammalien , Exome , Protéine du syndrome X fragile/métabolisme , Fréquence d'allèle , Génotype , Humains , Mâle , Retard mental lié à l'X/métabolisme , Retard mental lié à l'X/anatomopathologie , Souris , Protéines de tissu nerveux/déficit , Pedigree , Phénotype , Ténascine/déficit , Danio zébré
15.
Int J Cancer ; 149(5): 1100-1108, 2021 09 01.
Article de Anglais | MEDLINE | ID: mdl-33937989

RÉSUMÉ

Site-specific variation in colorectal cancer (CRC) incidence, biology and prognosis are poorly understood. We sought to determine whether common genetic variants influencing CRC risk might exhibit topographical differences on CRC risk through regional differences in effects on gene expression in the large bowel mucosa. We conducted a site-specific genetic association study (10 630 cases, 31 331 controls) to identify whether established risk variants exert differential effects on risk of proximal, compared to distal CRC. We collected normal colorectal mucosa and blood from 481 subjects and assessed mucosal gene expression using Illumina HumanHT-12v4 arrays in relation to germline genotype. Expression quantitative trait loci (eQTLs) were explored by anatomical location of sampling. The rs3087967 genotype (chr11q23.1 risk variant) exhibited significant site-specific effects-risk of distal CRC (odds ratio [OR] = 1.20, P = 8.20 × 10-20 ) with negligible effects on proximal CRC risk (OR = 1.05, P = .10). Expression of 1261 genes differed between proximal and distal colonic mucosa (top hit PRAC gene, fold-difference = 10, P = 3.48 × 10-57 ). In eQTL studies, rs3087967 genotype was associated with expression of 8 cis- and 21 trans-genes. Four of these (AKAP14, ADH5P4, ASGR2, RP11-342M1.7) showed differential effects by site, with strongest trans-eQTL signals in proximal colonic mucosa (eg, AKAP14, beta = 0.61, P = 5.02 × 10-5 ) and opposite signals in distal mucosa (AKAP14, beta = -0.17, P = .04). In summary, genetic variation at the chr11q23.1 risk locus imparts greater risk of distal rather than proximal CRC and exhibits site-specific differences in eQTL effects in normal mucosa. Topographical differences in genomic control over gene expression relevant to CRC risk may underlie site-specific variation in CRC. Results may inform individualised CRC screening programmes.


Sujet(s)
Marqueurs biologiques tumoraux/génétique , Tumeurs colorectales/génétique , Régulation de l'expression des gènes tumoraux , Prédisposition génétique à une maladie , Muqueuse intestinale/métabolisme , Polymorphisme de nucléotide simple , Locus de caractère quantitatif , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Études cas-témoins , Tumeurs colorectales/étiologie , Tumeurs colorectales/anatomopathologie , Femelle , Études de suivi , Étude d'association pangénomique , Génotype , Humains , Muqueuse intestinale/anatomopathologie , Mâle , Adulte d'âge moyen , Pronostic , Facteurs de risque , Transcriptome , Jeune adulte
16.
Nat Commun ; 12(1): 3127, 2021 05 25.
Article de Anglais | MEDLINE | ID: mdl-34035299

RÉSUMÉ

Cornelia de Lange syndrome is a multisystem developmental disorder typically caused by mutations in the gene encoding the cohesin loader NIPBL. The associated phenotype is generally assumed to be the consequence of aberrant transcriptional regulation. Recently, we identified a missense mutation in BRD4 associated with a Cornelia de Lange-like syndrome that reduces BRD4 binding to acetylated histones. Here we show that, although this mutation reduces BRD4-occupancy at enhancers it does not affect transcription of the pluripotency network in mouse embryonic stem cells. Rather, it delays the cell cycle, increases DNA damage signalling, and perturbs regulation of DNA repair in mutant cells. This uncovers a role for BRD4 in DNA repair pathway choice. Furthermore, we find evidence of a similar increase in DNA damage signalling in cells derived from NIPBL-deficient individuals, suggesting that defective DNA damage signalling and repair is also a feature of typical Cornelia de Lange syndrome.


Sujet(s)
Altération de l'ADN , Réparation de l'ADN , Syndrome de Cornelia de Lange/génétique , Mutation , Animaux , Protéines du cycle cellulaire/génétique , Lignée cellulaire , Lignée cellulaire tumorale , Cellules cultivées , Prédisposition génétique à une maladie/génétique , Humains , Souris , RNA-Seq/méthodes , Transduction du signal/génétique , Facteurs de transcription/génétique
17.
Clin Cancer Res ; 27(11): 3201-3214, 2021 06 01.
Article de Anglais | MEDLINE | ID: mdl-33741650

RÉSUMÉ

PURPOSE: The abundance and effects of structural variation at BRCA1/2 in tumors are not well understood. In particular, the impact of these events on homologous recombination repair deficiency (HRD) has yet to be demonstrated. EXPERIMENTAL DESIGN: Exploiting a large collection of whole-genome sequencing data from high-grade serous ovarian carcinoma (N = 205) together with matched RNA sequencing for the majority of tumors (N = 150), we have comprehensively characterized mutation and expression at BRCA1/2. RESULTS: In addition to the known spectrum of short somatic mutations (SSM), we discovered that multi-megabase structural variants (SV) were a frequent, unappreciated source of BRCA1/2 disruption in these tumors, and we found a genome-wide enrichment for large deletions at the BRCA1/2 loci across the cohort. These SVs independently affected a substantial proportion of patients (16%) in addition to those affected by SSMs (24%), conferring HRD and impacting patient survival. We also detail compound deficiencies involving SSMs and SVs at both loci, demonstrating that the strongest risk of HRD emerges from combined SVs at both BRCA1 and BRCA2 in the absence of SSMs. Furthermore, these SVs are abundant and disruptive in other cancer types. CONCLUSIONS: These results extend our understanding of the mutational landscape underlying HRD, increase the number of patients predicted to benefit from therapies exploiting HRD, and suggest there is currently untapped potential in SV detection for patient stratification.


Sujet(s)
Protéine BRCA1/génétique , Protéine BRCA2/génétique , Cystadénocarcinome séreux/génétique , Cystadénocarcinome séreux/anatomopathologie , Recombinaison homologue/génétique , Mutation/génétique , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Réparation de l'ADN par recombinaison/génétique , Protéine BRCA1/métabolisme , Protéine BRCA2/métabolisme , Femelle , Expression des gènes , Humains , Séquençage du génome entier
18.
Sci Rep ; 11(1): 229, 2021 01 08.
Article de Anglais | MEDLINE | ID: mdl-33420223

RÉSUMÉ

Focal adhesion kinase (FAK) localizes to focal adhesions and is overexpressed in many cancers. FAK can also translocate to the nucleus, where it binds to, and regulates, several transcription factors, including MBD2, p53 and IL-33, to control gene expression by unknown mechanisms. We have used ATAC-seq to reveal that FAK controls chromatin accessibility at a subset of regulated genes. Integration of ATAC-seq and RNA-seq data showed that FAK-dependent chromatin accessibility is linked to differential gene expression, including of the FAK-regulated cytokine and transcriptional regulator interleukin-33 (Il33), which controls anti-tumor immunity. Analysis of the accessibility peaks on the Il33 gene promoter/enhancer regions revealed sequences for several transcription factors, including ETS and AP-1 motifs, and we show that c-Jun, a component of AP-1, regulates Il33 gene expression by binding to its enhancer in a FAK kinase-dependent manner. This work provides the first demonstration that FAK controls transcription via chromatin accessibility, identifying a novel mechanism by which nuclear FAK regulates biologically important gene expression.


Sujet(s)
Chromatine/métabolisme , Focal adhesion protein-tyrosine kinases/métabolisme , Régulation de l'expression des gènes , Interleukine-33/métabolisme , JNK Mitogen-Activated Protein Kinases/métabolisme , Motifs d'acides aminés , Communication cellulaire , Noyau de la cellule/métabolisme , Humains , Liaison aux protéines
19.
Nature ; 591(7848): 92-98, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-33307546

RÉSUMÉ

Host-mediated lung inflammation is present1, and drives mortality2, in the critical illness caused by coronavirus disease 2019 (COVID-19). Host genetic variants associated with critical illness may identify mechanistic targets for therapeutic development3. Here we report the results of the GenOMICC (Genetics Of Mortality In Critical Care) genome-wide association study in 2,244 critically ill patients with COVID-19 from 208 UK intensive care units. We have identified and replicated the following new genome-wide significant associations: on chromosome 12q24.13 (rs10735079, P = 1.65 × 10-8) in a gene cluster that encodes antiviral restriction enzyme activators (OAS1, OAS2 and OAS3); on chromosome 19p13.2 (rs74956615, P = 2.3 × 10-8) near the gene that encodes tyrosine kinase 2 (TYK2); on chromosome 19p13.3 (rs2109069, P = 3.98 ×  10-12) within the gene that encodes dipeptidyl peptidase 9 (DPP9); and on chromosome 21q22.1 (rs2236757, P = 4.99 × 10-8) in the interferon receptor gene IFNAR2. We identified potential targets for repurposing of licensed medications: using Mendelian randomization, we found evidence that low expression of IFNAR2, or high expression of TYK2, are associated with life-threatening disease; and transcriptome-wide association in lung tissue revealed that high expression of the monocyte-macrophage chemotactic receptor CCR2 is associated with severe COVID-19. Our results identify robust genetic signals relating to key host antiviral defence mechanisms and mediators of inflammatory organ damage in COVID-19. Both mechanisms may be amenable to targeted treatment with existing drugs. However, large-scale randomized clinical trials will be essential before any change to clinical practice.


Sujet(s)
COVID-19/génétique , COVID-19/physiopathologie , Maladie grave , 2',5'-Oligoadenylate synthetase/génétique , COVID-19/anatomopathologie , Chromosomes humains de la paire 12/génétique , Chromosomes humains de la paire 19/génétique , Chromosomes humains de la paire 21/génétique , Soins de réanimation , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/génétique , Repositionnement des médicaments , Femelle , Étude d'association pangénomique , Humains , Inflammation/génétique , Inflammation/anatomopathologie , Inflammation/physiopathologie , Poumon/anatomopathologie , Poumon/physiopathologie , Poumon/virologie , Mâle , Famille multigénique/génétique , Récepteur à l'interféron alpha-bêta/génétique , Récepteurs CCR2/génétique , TYK2 Kinase/génétique , Royaume-Uni
20.
Cell Rep ; 29(7): 1974-1985.e6, 2019 11 12.
Article de Anglais | MEDLINE | ID: mdl-31722211

RÉSUMÉ

The DNA hypomethylation that occurs when embryonic stem cells (ESCs) are directed to the ground state of naive pluripotency by culturing in two small molecule inhibitors (2i) results in redistribution of polycomb (H3K27me3) away from its target loci. Here, we demonstrate that 3D genome organization is also altered in 2i, with chromatin decompaction at polycomb target loci and a loss of long-range polycomb interactions. By preventing DNA hypomethylation during the transition to the ground state, we are able to restore to ESC in 2i the H3K27me3 distribution, as well as polycomb-mediated 3D genome organization that is characteristic of primed ESCs grown in serum. However, these cells retain the functional characteristics of 2i ground-state ESCs. Our findings demonstrate the central role of DNA methylation in shaping major aspects of 3D genome organization but caution against assuming causal roles for the epigenome and 3D genome in gene regulation and function in ESCs.


Sujet(s)
Assemblage et désassemblage de la chromatine , Chromatine/métabolisme , Méthylation de l'ADN , Épigénome , Cellules souches embryonnaires de souris/métabolisme , Animaux , Chromatine/génétique , Mâle , Souris , Souris knockout , Cellules souches embryonnaires de souris/cytologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE