Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 80
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
bioRxiv ; 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39229041

RÉSUMÉ

Epigenetic dysregulation is widespread in cancer. However, the specific epigenetic regulators and the processes they control to drive cancer phenotypes are poorly understood. Here, we employed a novel, scalable and high-throughput in vivo method to perform iterative functional screens of over 250 epigenetic regulatory genes within autochthonous oncogenic KRAS-driven lung tumors. We identified multiple novel epigenetic tumor suppressor and tumor dependency genes. We show that a specific HBO1 complex and the MLL1 complex are among the most impactful tumor suppressive epigenetic regulators in lung. The histone modifications generated by the HBO1 complex are frequently absent or reduced in human lung adenocarcinomas. The HBO1 and MLL1 complexes regulate chromatin accessibility of shared genomic regions, lineage fidelity and the expression of canonical tumor suppressor genes. The HBO1 and MLL1 complexes are epistatic during lung tumorigenesis, and their functional correlation is conserved in human cancer cell lines. Together, these results demonstrate the value of quantitative methods to generate a phenotypic roadmap of epigenetic regulatory genes in tumorigenesis in vivo .

2.
J Clin Invest ; 134(7)2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38557491

RÉSUMÉ

Mutations in genes encoding chromatin modifiers are enriched among mutations causing intellectual disability. The continuing development of the brain postnatally, coupled with the inherent reversibility of chromatin modifications, may afford an opportunity for therapeutic intervention following a genetic diagnosis. Development of treatments requires an understanding of protein function and models of the disease. Here, we provide a mouse model of Say-Barber-Biesecker-Young-Simpson syndrome (SBBYSS) (OMIM 603736) and demonstrate proof-of-principle efficacy of postnatal treatment. SBBYSS results from heterozygous mutations in the KAT6B (MYST4/MORF/QFK) gene and is characterized by intellectual disability and autism-like behaviors. Using human cells carrying SBBYSS-specific KAT6B mutations and Kat6b heterozygous mice (Kat6b+/-), we showed that KAT6B deficiency caused a reduction in histone H3 lysine 9 acetylation. Kat6b+/- mice displayed learning, memory, and social deficits, mirroring SBBYSS individuals. Treatment with a histone deacetylase inhibitor, valproic acid, or an acetyl donor, acetyl-carnitine (ALCAR), elevated histone acetylation levels in the human cells with SBBYSS mutations and in brain and blood cells of Kat6b+/- mice and partially reversed gene expression changes in Kat6b+/- cortical neurons. Both compounds improved sociability in Kat6b+/- mice, and ALCAR treatment restored learning and memory. These data suggest that a subset of SBBYSS individuals may benefit from postnatal therapeutic interventions.


Sujet(s)
Malformations multiples , Acétyl-carnitine , Hypothyroïdie congénitale , Malformations crâniofaciales , Histone acetyltransferases , Déficience intellectuelle , Instabilité articulaire , Animaux , Humains , Souris , Malformations multiples/traitement médicamenteux , Malformations multiples/génétique , Acétylation , Acétyl-carnitine/pharmacologie , Acétyl-carnitine/usage thérapeutique , Blépharophimosis , Chromatine , Malformations crâniofaciales/traitement médicamenteux , Malformations crâniofaciales/génétique , Exons , Faciès , Cardiopathies congénitales , Histone acetyltransferases/antagonistes et inhibiteurs , Histone acetyltransferases/génétique , Histone acetyltransferases/métabolisme , Histone/génétique , Déficience intellectuelle/traitement médicamenteux , Déficience intellectuelle/génétique
3.
Development ; 151(5)2024 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-38446206

RÉSUMÉ

Inhibitor of growth 4 and 5 (ING4, ING5) are structurally similar chromatin-binding proteins in the KAT6A, KAT6B and KAT7 histone acetyltransferase protein complexes. Heterozygous mutations in the KAT6A or KAT6B gene cause human disorders with cardiac defects, but the contribution of their chromatin-adaptor proteins to development is unknown. We found that Ing5-/- mice had isolated cardiac ventricular septal defects. Ing4-/-Ing5-/- embryos failed to undergo chorioallantoic fusion and arrested in development at embryonic day 8.5, displaying loss of histone H3 lysine 14 acetylation, reduction in H3 lysine 23 acetylation levels and reduced developmental gene expression. Embryonic day 12.5 Ing4+/-Ing5-/- hearts showed a paucity of epicardial cells and epicardium-derived cells, failure of myocardium compaction, and coronary vasculature defects, accompanied by reduced expression of epicardium genes. Cell adhesion gene expression and proepicardium outgrowth were defective in the ING4- and ING5-deficient state. Our findings suggest that ING4 and ING5 are essential for heart development and promote epicardium and epicardium-derived cell fates and imply mutation of the human ING5 gene as a possible cause of isolated ventricular septal defects.


Sujet(s)
Protéines de transport , Communications interventriculaires , Lysine , Humains , Animaux , Souris , Lignage cellulaire , Histone , Acétylation , Chromatine , Facteurs de transcription , Protéines suppresseurs de tumeurs , Protéines à homéodomaine/génétique , Protéines du cycle cellulaire , Histone acetyltransferases
4.
EMBO Rep ; 25(3): 1256-1281, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38429579

RÉSUMÉ

The plant homeodomain zinc-finger protein, PHF6, is a transcriptional regulator, and PHF6 germline mutations cause the X-linked intellectual disability (XLID) Börjeson-Forssman-Lehmann syndrome (BFLS). The mechanisms by which PHF6 regulates transcription and how its mutations cause BFLS remain poorly characterized. Here, we show genome-wide binding of PHF6 in the developing cortex in the vicinity of genes involved in central nervous system development and neurogenesis. Characterization of BFLS mice harbouring PHF6 patient mutations reveals an increase in embryonic neural stem cell (eNSC) self-renewal and a reduction of neural progenitors. We identify a panel of Ephrin receptors (EphRs) as direct transcriptional targets of PHF6. Mechanistically, we show that PHF6 regulation of EphR is impaired in BFLS mice and in conditional Phf6 knock-out mice. Knockdown of EphR-A phenocopies the PHF6 loss-of-function defects in altering eNSCs, and its forced expression rescues defects of BFLS mice-derived eNSCs. Our data indicate that PHF6 directly promotes Ephrin receptor expression to control eNSC behaviour in the developing brain, and that this pathway is impaired in BFLS.


Sujet(s)
Épilepsie , Face/malformations , Doigts/malformations , Troubles de la croissance , Hypogonadisme , Déficience intellectuelle , Retard mental lié à l'X , Obésité , Humains , Souris , Animaux , Déficience intellectuelle/génétique , Protéines de répression , Retard mental lié à l'X/génétique , Retard mental lié à l'X/métabolisme , Épilepsie/génétique , Épilepsie/métabolisme , Facteurs de transcription
5.
Mol Genet Metab ; 142(1): 108360, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38428378

RÉSUMÉ

The Mendelian disorders of chromatin machinery (MDCMs) represent a distinct subgroup of disorders that present with neurodevelopmental disability. The chromatin machinery regulates gene expression by a range of mechanisms, including by post-translational modification of histones, responding to histone marks, and remodelling nucleosomes. Some of the MDCMs that impact on histone modification may have potential therapeutic interventions. Two potential treatment strategies are to enhance the intracellular pool of metabolites that can act as substrates for histone modifiers and the use of medications that may inhibit or promote the modification of histone residues to influence gene expression. In this article we discuss the influence and potential treatments of histone modifications involving histone acetylation and histone methylation. Genomic technologies are facilitating earlier diagnosis of many Mendelian disorders, providing potential opportunities for early treatment from infancy. This has parallels with how inborn errors of metabolism have been afforded early treatment with newborn screening. Before this promise can be fulfilled, we require greater understanding of the biochemical fingerprint of these conditions, which may provide opportunities to supplement metabolites that can act as substrates for chromatin modifying enzymes. Importantly, understanding the metabolomic profile of affected individuals may also provide disorder-specific biomarkers that will be critical for demonstrating efficacy of treatment, as treatment response may not be able to be accurately assessed by clinical measures.


Sujet(s)
Chromatine , Voies et réseaux métaboliques , Humains , Chromatine/génétique , Chromatine/métabolisme , Voies et réseaux métaboliques/génétique , Histone/métabolisme , Histone/génétique , Maturation post-traductionnelle des protéines , Acétylation , Erreurs innées du métabolisme/génétique , Erreurs innées du métabolisme/thérapie , Erreurs innées du métabolisme/diagnostic , Erreurs innées du métabolisme/métabolisme , Assemblage et désassemblage de la chromatine/génétique , Maladies génétiques congénitales/génétique , Maladies génétiques congénitales/thérapie , Maladies génétiques congénitales/métabolisme , Nouveau-né , Méthylation
6.
Stem Cell Reports ; 19(4): 469-485, 2024 Apr 09.
Article de Anglais | MEDLINE | ID: mdl-38518784

RÉSUMÉ

The histone lysine acetyltransferase KAT6B (MYST4, MORF, QKF) is the target of recurrent chromosomal translocations causing hematological malignancies with poor prognosis. Using Kat6b germline deletion and overexpression in mice, we determined the role of KAT6B in the hematopoietic system. We found that KAT6B sustained the fetal hematopoietic stem cell pool but did not affect viability or differentiation. KAT6B was essential for normal levels of histone H3 lysine 9 (H3K9) acetylation but not for a previously proposed target, H3K23. Compound heterozygosity of Kat6b and the closely related gene, Kat6a, abolished hematopoietic reconstitution after transplantation. KAT6B and KAT6A cooperatively promoted transcription of genes regulating hematopoiesis, including the Hoxa cluster, Pbx1, Meis1, Gata family, Erg, and Flt3. In conclusion, we identified the hematopoietic processes requiring Kat6b and showed that KAT6B and KAT6A synergistically promoted HSC development, function, and transcription. Our findings are pertinent to current clinical trials testing KAT6A/B inhibitors as cancer therapeutics.


Sujet(s)
Tumeurs hématologiques , Hématopoïèse , Souris , Animaux , Différenciation cellulaire/génétique , Cellules souches hématopoïétiques , Histone acetyltransferases/génétique
7.
Cell Death Differ ; 31(2): 150-158, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-38097622

RÉSUMÉ

Whole-genome screens using CRISPR technologies are powerful tools to identify novel tumour suppressors as well as factors that impact responses of malignant cells to anti-cancer agents. Applying this methodology to lymphoma cells, we conducted a genome-wide screen to identify novel inhibitors of tumour expansion that are induced by the tumour suppressor TRP53. We discovered that the absence of Arrestin domain containing 3 (ARRDC3) increases the survival and long-term competitiveness of MYC-driven lymphoma cells when treated with anti-cancer agents that activate TRP53. Deleting Arrdc3 in mice caused perinatal lethality due to various developmental abnormalities, including cardiac defects. Notably, the absence of ARRDC3 markedly accelerated MYC-driven lymphoma development. Thus, ARRDC3 is a new mediator of TRP53-mediated suppression of tumour expansion, and this discovery may open new avenues to harness this process for cancer therapy.


Sujet(s)
Lymphomes , Tumeurs , Animaux , Souris , Arrestines/génétique , Arrestines/métabolisme , Clustered regularly interspaced short palindromic repeats , Tumeurs/génétique
8.
Front Immunol ; 14: 1119750, 2023.
Article de Anglais | MEDLINE | ID: mdl-37275850

RÉSUMÉ

ING5 is a component of KAT6A and KAT7 histone lysine acetylation protein complexes. ING5 contains a PHD domain that binds to histone H3 lysine 4 when it is trimethylated, and so functions as a 'reader' and adaptor protein. KAT6A and KAT7 function are critical for normal hematopoiesis. To examine the function of ING5 in hematopoiesis, we generated a null allele of Ing5. Mice lacking ING5 during development had decreased foetal liver cellularity, decreased numbers of hematopoietic stem cells and perturbed erythropoiesis compared to wild-type control mice. Ing5-/- pups had hypoplastic spleens. Competitive transplantation experiments using foetal liver hematopoietic cells showed that there was no defect in long-term repopulating capacity of stem cells lacking ING5, suggesting that the defects during the foetal stage were not cell intrinsic. Together, these results suggest that ING5 function is dispensable for normal hematopoiesis but may be required for timely foetal hematopoiesis in a cell-extrinsic manner.


Sujet(s)
Transplantation de cellules souches hématopoïétiques , Facteurs de transcription , Protéines suppresseurs de tumeurs , Animaux , Souris , Numération cellulaire , Chromatine/génétique , Foie , Lysine , Protéines suppresseurs de tumeurs/génétique , Facteurs de transcription/génétique
9.
Cell Rep ; 42(1): 111980, 2023 01 31.
Article de Anglais | MEDLINE | ID: mdl-36641753

RÉSUMÉ

In the conventional model of transcriptional activation, transcription factors bind to response elements and recruit co-factors, including histone acetyltransferases. Contrary to this model, we show that the histone acetyltransferase KAT7 (HBO1/MYST2) is required genome wide for histone H3 lysine 14 acetylation (H3K14ac). Examining neural stem cells, we find that KAT7 and H3K14ac are present not only at transcribed genes but also at inactive genes, intergenic regions, and in heterochromatin. KAT7 and H3K14ac were not required for the continued transcription of genes that were actively transcribed at the time of loss of KAT7 but indispensable for the activation of repressed genes. The absence of KAT7 abrogates neural stem cell plasticity, diverse differentiation pathways, and cerebral cortex development. Re-expression of KAT7 restored stem cell developmental potential. Overexpression of KAT7 enhanced neuron and oligodendrocyte differentiation. Our data suggest that KAT7 prepares chromatin for transcriptional activation and is a prerequisite for gene activation.


Sujet(s)
Plasticité cellulaire , Histone , Histone/métabolisme , Activation de la transcription/génétique , Acétylation , Plasticité cellulaire/génétique , Cellules souches/métabolisme , Histone acetyltransferases/génétique , Histone acetyltransferases/métabolisme
10.
Cell Death Dis ; 13(7): 627, 2022 07 20.
Article de Anglais | MEDLINE | ID: mdl-35853868

RÉSUMÉ

Histone acetylation is essential for initiating and maintaining a permissive chromatin conformation and gene transcription. Dysregulation of histone acetylation can contribute to tumorigenesis and metastasis. Using inducible cre-recombinase and CRISPR/Cas9-mediated deletion, we investigated the roles of the histone lysine acetyltransferase TIP60 (KAT5/HTATIP) in human cells, mouse cells, and mouse embryos. We found that loss of TIP60 caused complete cell growth arrest. In the absence of TIP60, chromosomes failed to align in a metaphase plate during mitosis. In some TIP60 deleted cells, endoreplication occurred instead. In contrast, cell survival was not affected. Remarkably, the cell growth arrest caused by loss of TIP60 was independent of the tumor suppressors p53, INK4A and ARF. TIP60 was found to be essential for the acetylation of H2AZ, specifically at lysine 7. The mRNA levels of 6236 human and 8238 mouse genes, including many metabolism genes, were dependent on TIP60. Among the top 50 differentially expressed genes, over 90% were downregulated in cells lacking TIP60, supporting a role for TIP60 as a key co-activator of transcription. We propose a primary role of TIP60 in H2AZ lysine 7 acetylation and transcriptional activation, and that this fundamental role is essential for cell proliferation. Growth arrest independent of major tumor suppressors suggests TIP60 as a potential anti-cancer drug target.


Sujet(s)
Histone , Lysine acetyltransferase 5 , Lysine , Protéine p53 suppresseur de tumeur , Acétylation , Animaux , Points de contrôle du cycle cellulaire/physiologie , Inhibiteur p16 de kinase cycline-dépendante/génétique , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Histone acetyltransferases/génétique , Histone acetyltransferases/métabolisme , Histone/génétique , Histone/métabolisme , Humains , Lysine/métabolisme , Lysine acetyltransferase 5/déficit , Lysine acetyltransferase 5/génétique , Lysine acetyltransferase 5/métabolisme , Souris , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme
11.
EMBO J ; 41(15): e110300, 2022 08 01.
Article de Anglais | MEDLINE | ID: mdl-35758142

RÉSUMÉ

The intrinsic apoptosis pathway, regulated by the BCL-2 protein family, is essential for embryonic development. Using mice lacking all known apoptosis effectors, BAX, BAK and BOK, we have previously defined the processes during development that require apoptosis. Rare Bok-/- Bax-/- Bak-/- triple knockout (TKO) mice developed to adulthood and several tissues that were thought to require apoptosis during development appeared normal. This raises the question if all apoptosis had been abolished in the TKO mice or if other BCL-2 family members could act as effectors of apoptosis. Here, we investigated the role of BID, generally considered to link the extrinsic and intrinsic apoptosis pathways, acting as a BH3-only protein initiating apoptosis upstream of BAX and BAK. We found that Bok-/- Bax-/- Bak-/- Bid-/- quadruple knockout (QKO) mice have additional developmental anomalies compared to TKO mice, consistent with a role of BID, not only upstream but also in parallel to BAX, BAK and BOK. Mitochondrial experiments identified a small cytochrome c-releasing activity of full-length BID. Collectively, these findings suggest a new effector role for BID in the intrinsic apoptosis pathway.


Sujet(s)
Protéine Bid , Protéines proto-oncogènes c-bcl-2 , Protéine Bak , Animaux , Souris , Apoptose , Protéine Bak/génétique , Protéine Bak/métabolisme , Protéine Bax/génétique , Protéine Bax/métabolisme , Protéine Bid/génétique , Protéine Bid/métabolisme , Développement embryonnaire/génétique , Souris knockout , Protéines proto-oncogènes c-bcl-2/génétique , Protéines proto-oncogènes c-bcl-2/métabolisme
13.
Cell Death Differ ; 29(5): 1013-1027, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-35361962

RÉSUMÉ

Mutations in genes encoding general transcription factors cause neurological disorders. Despite clinical prominence, the consequences of defects in the basal transcription machinery during brain development are unclear. We found that loss of the TATA-box binding protein-associated factor TAF8, a component of the general transcription factor TFIID, in the developing central nervous system affected the expression of many, but notably not all genes. Taf8 deletion caused apoptosis, unexpectedly restricted to forebrain regions. Nuclear levels of the transcription factor p53 were elevated in the absence of TAF8, as were the mRNAs of the pro-apoptotic p53 target genes Noxa, Puma and Bax. The cell death in Taf8 forebrain regions was completely rescued by additional loss of p53, but Taf8 and p53 brains failed to initiate a neuronal expression program. Taf8 deletion caused aberrant transcription of promoter regions and splicing anomalies. We propose that TAF8 supports the directionality of transcription and co-transcriptional splicing, and that failure of these processes causes p53-induced apoptosis of neuronal cells in the developing mouse embryo.


Sujet(s)
Facteur de transcription TFIID , Facteurs de transcription/métabolisme , Protéine p53 suppresseur de tumeur , Animaux , Apoptose/génétique , Mort cellulaire , Souris , Facteur de transcription TFIID/génétique , Facteur de transcription TFIID/métabolisme , Transcription génétique , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme
14.
Sci Immunol ; 7(67): eabb6032, 2022 01 21.
Article de Anglais | MEDLINE | ID: mdl-35061506

RÉSUMÉ

The autoimmune regulator (AIRE) induces the transcription of thousands of peripheral tissue genes (PTGs) in thymic epithelial cells (TECs) to mediate immunological tolerance. The chromatin state required for optimal AIRE function in TECs and how this state is induced remains unclear. We tested the role of the histone acetyltransferase, KAT7 (also known as HBO1 or MYST2), which is essential for acetylation of histone 3 lysine 14, in TEC differentiation, AIRE-mediated PTG expression, and thymic tolerance. We find that KAT7 is required for optimal expansion of medullary TEC and has a major role in the expression of AIRE-dependent PTGs, associated with enhanced chromatin accessibility at these gene loci in TECs. Mice with TEC-specific Kat7 deletion develop organ-specific autoimmunity with features resembling those observed in Aire-deficient mice. These findings highlight critical roles for KAT7-mediated acetylation in promoting a chromatin state at PTG loci that enables AIRE function and the establishment of immunological tolerance.


Sujet(s)
Cellules épithéliales/immunologie , Histone acetyltransferases/immunologie , Thymus (glande)/immunologie , Facteurs de transcription/immunologie , Animaux , Tolérance immunitaire/immunologie , Souris , Souris de lignée C57BL , Souris knockout , Souris transgéniques , Thymus (glande)/cytologie ,
15.
Blood ; 139(6): 845-858, 2022 02 10.
Article de Anglais | MEDLINE | ID: mdl-34724565

RÉSUMÉ

The histone acetyltransferase HBO1 (MYST2, KAT7) is indispensable for postgastrulation development, histone H3 lysine 14 acetylation (H3K14Ac), and the expression of embryonic patterning genes. In this study, we report the role of HBO1 in regulating hematopoietic stem cell function in adult hematopoiesis. We used 2 complementary cre-recombinase transgenes to conditionally delete Hbo1 (Mx1-Cre and Rosa26-CreERT2). Hbo1-null mice became moribund due to hematopoietic failure with pancytopenia in the blood and bone marrow 2 to 6 weeks after Hbo1 deletion. Hbo1-deleted bone marrow cells failed to repopulate hemoablated recipients in competitive transplantation experiments. Hbo1 deletion caused a rapid loss of hematopoietic progenitors. The numbers of lineage-restricted progenitors for the erythroid, myeloid, B-, and T-cell lineages were reduced. Loss of HBO1 resulted in an abnormally high rate of recruitment of quiescent hematopoietic stem cells (HSCs) into the cell cycle. Cycling HSCs produced progenitors at the expense of self-renewal, which led to the exhaustion of the HSC pool. Mechanistically, genes important for HSC functions were downregulated in HSC-enriched cell populations after Hbo1 deletion, including genes essential for HSC quiescence and self-renewal, such as Mpl, Tek(Tie-2), Gfi1b, Egr1, Tal1(Scl), Gata2, Erg, Pbx1, Meis1, and Hox9, as well as genes important for multipotent progenitor cells and lineage-specific progenitor cells, such as Gata1. HBO1 was required for H3K14Ac through the genome and particularly at gene loci required for HSC quiescence and self-renewal. Our data indicate that HBO1 promotes the expression of a transcription factor network essential for HSC maintenance and self-renewal in adult hematopoiesis.


Sujet(s)
Auto-renouvellement cellulaire , Cellules souches hématopoïétiques , Histone acetyltransferases , Animaux , Cellules cultivées , Vieillissement de la cellule , Délétion de gène , Hématopoïèse , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/métabolisme , Histone acetyltransferases/génétique , Histone acetyltransferases/métabolisme , Souris de lignée C57BL
17.
Development ; 148(20)2021 10 15.
Article de Anglais | MEDLINE | ID: mdl-34550360

RÉSUMÉ

Blood vessel growth and remodelling are essential during embryonic development and disease pathogenesis. The diversity of endothelial cells (ECs) is transcriptionally evident and ECs undergo dynamic changes in gene expression during vessel growth and remodelling. Here, we investigated the role of the histone acetyltransferase HBO1 (KAT7), which is important for activating genes during development and for histone H3 lysine 14 acetylation (H3K14ac). Loss of HBO1 and H3K14ac impaired developmental sprouting angiogenesis and reduced pathological EC overgrowth in the retinal endothelium. Single-cell RNA sequencing of retinal ECs revealed an increased abundance of tip cells in Hbo1-deficient retinas, which led to EC overcrowding in the retinal sprouting front and prevented efficient tip cell migration. We found that H3K14ac was highly abundant in the endothelial genome in both intra- and intergenic regions, suggesting that HBO1 acts as a genome organiser that promotes efficient tip cell behaviour necessary for sprouting angiogenesis. This article has an associated 'The people behind the papers' interview.


Sujet(s)
Histone acetyltransferases/métabolisme , Néovascularisation pathologique/métabolisme , Acétylation , Animaux , Mouvement cellulaire/physiologie , Cellules cultivées , Développement embryonnaire/physiologie , Cellules endothéliales/métabolisme , Femelle , Histone/métabolisme , Cellules endothéliales de la veine ombilicale humaine , Humains , Lysine/métabolisme , Souris , Souris de lignée BALB C , Souris de lignée C57BL
18.
Development ; 147(21)2020 10 23.
Article de Anglais | MEDLINE | ID: mdl-32994169

RÉSUMÉ

Börjeson-Forssman-Lehmann syndrome (BFLS) is an intellectual disability and endocrine disorder caused by plant homeodomain finger 6 (PHF6) mutations. Individuals with BFLS present with short stature. We report a mouse model of BFLS, in which deletion of Phf6 causes a proportional reduction in body size compared with control mice. Growth hormone (GH) levels were reduced in the absence of PHF6. Phf6-/Y animals displayed a reduction in the expression of the genes encoding GH-releasing hormone (GHRH) in the brain, GH in the pituitary gland and insulin-like growth factor 1 (IGF1) in the liver. Phf6 deletion specifically in the nervous system caused a proportional growth defect, indicating a neuroendocrine contribution to the phenotype. Loss of suppressor of cytokine signaling 2 (SOCS2), a negative regulator of growth hormone signaling partially rescued body size, supporting a reversible deficiency in GH signaling. These results demonstrate that PHF6 regulates the GHRH/GH/IGF1 axis.


Sujet(s)
Régulation négative , Épilepsie/métabolisme , Face/malformations , Doigts/malformations , Troubles de la croissance/métabolisme , Hormone de libération de l'hormone de croissance/métabolisme , Hormone de croissance/métabolisme , Hypogonadisme/métabolisme , Facteur de croissance IGF-I/métabolisme , Retard mental lié à l'X/métabolisme , Obésité/métabolisme , Protéines de répression/métabolisme , Transduction du signal , Animaux , Animaux nouveau-nés , Modèles animaux de maladie humaine , Épilepsie/sang , Épilepsie/anatomopathologie , Face/anatomopathologie , Doigts/anatomopathologie , Troubles de la croissance/sang , Troubles de la croissance/anatomopathologie , Hormone de croissance/sang , Hypogonadisme/sang , Hypogonadisme/anatomopathologie , Hypothalamus/métabolisme , Facteur de croissance IGF-I/génétique , Mâle , Retard mental lié à l'X/sang , Retard mental lié à l'X/anatomopathologie , Souris , Souris de lignée C57BL , Souris knockout , Système nerveux/métabolisme , Obésité/sang , Obésité/anatomopathologie , Spécificité d'organe , Hypophyse/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Protéines SOCS/métabolisme
19.
J Med Chem ; 63(9): 4655-4684, 2020 05 14.
Article de Anglais | MEDLINE | ID: mdl-32118427

RÉSUMÉ

A high-throughput screen designed to discover new inhibitors of histone acetyltransferase KAT6A uncovered CTX-0124143 (1), a unique aryl acylsulfonohydrazide with an IC50 of 1.0 µM. Using this acylsulfonohydrazide as a template, we herein disclose the results of our extensive structure-activity relationship investigations, which resulted in the discovery of advanced compounds such as 55 and 80. These two compounds represent significant improvements on our recently reported prototypical lead WM-8014 (3) as they are not only equivalently potent as inhibitors of KAT6A but are less lipophilic and significantly more stable to microsomal degradation. Furthermore, during this process, we discovered a distinct structural subclass that contains key 2-fluorobenzenesulfonyl and phenylpyridine motifs, culminating in the discovery of WM-1119 (4). This compound is a highly potent KAT6A inhibitor (IC50 = 6.3 nM; KD = 0.002 µM), competes with Ac-CoA by binding to the Ac-CoA binding site, and has an oral bioavailability of 56% in rats.


Sujet(s)
Antinéoplasiques/pharmacologie , Histone acetyltransferases/antagonistes et inhibiteurs , Hydrazines/pharmacologie , Sulfonamides/pharmacologie , Animaux , Antinéoplasiques/synthèse chimique , Antinéoplasiques/pharmacocinétique , Biodisponibilité , Découverte de médicament , Stabilité de médicament , Humains , Hydrazines/synthèse chimique , Hydrazines/composition chimique , Hydrazines/pharmacocinétique , Mâle , Souris , Microsomes du foie/métabolisme , Structure moléculaire , Rat Sprague-Dawley , Relation structure-activité , Sulfonamides/synthèse chimique , Sulfonamides/composition chimique , Sulfonamides/métabolisme , Sulfonamides/pharmacocinétique
20.
Cell Rep ; 30(11): 3717-3728.e6, 2020 03 17.
Article de Anglais | MEDLINE | ID: mdl-32187544

RÉSUMÉ

Understanding the mechanisms of activity-dependent gene transcription underlying adaptive behaviors is challenging at neuronal-subtype resolution. Using cell-type specific molecular analysis in agouti-related peptide (AgRP) neurons, we reveal that the profound hunger-induced transcriptional changes greatly depend on plant homeodomain finger protein 6 (PHF6), a transcriptional repressor enriched in AgRP neurons. Loss of PHF6 in the satiated mice results in a hunger-state-shifting transcriptional profile, while hunger fails to further induce a rapid and robust activity-dependent gene transcription in PHF6-deficient AgRP neurons. We reveal that PHF6 binds to the promoters of a subset of immediate-early genes (IEGs) and that this chromatin binding is dynamically regulated by hunger state. Depletion of PHF6 decreases hunger-driven feeding motivation and makes the mice resistant to body weight gain under repetitive fasting-refeeding conditions. Our work identifies a neuronal subtype-specific transcriptional repressor that modulates transcriptional profiles in different nutritional states and enables adaptive eating behavior.


Sujet(s)
Chromatine/métabolisme , Réseaux de régulation génique/génétique , Faim/physiologie , Neurones/métabolisme , Protéines de répression/métabolisme , Protéine apparentée à Agouti/métabolisme , Animaux , Régime alimentaire , Régulation négative/génétique , Comportement alimentaire , Gene Ontology , Gènes précoces , Hypothalamus/métabolisme , Souris de lignée C57BL , Motivation , Régions promotrices (génétique)/génétique , Liaison aux protéines , Protéines de répression/génétique , Sensation de satiété , Prise de poids
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE