Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Clin Invest ; 134(7)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38557491

RESUMEN

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.


Asunto(s)
Anomalías Múltiples , Acetilcarnitina , Hipotiroidismo Congénito , Anomalías Craneofaciales , Histona Acetiltransferasas , Discapacidad Intelectual , Inestabilidad de la Articulación , Animales , Humanos , Ratones , Anomalías Múltiples/tratamiento farmacológico , Anomalías Múltiples/genética , Acetilación , Acetilcarnitina/farmacología , Acetilcarnitina/uso terapéutico , Blefarofimosis , Cromatina , Anomalías Craneofaciales/tratamiento farmacológico , Anomalías Craneofaciales/genética , Exones , Facies , Cardiopatías Congénitas , Histona Acetiltransferasas/antagonistas & inhibidores , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Histonas/genética , Discapacidad Intelectual/tratamiento farmacológico , Discapacidad Intelectual/genética
2.
Development ; 151(5)2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38446206

RESUMEN

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.


Asunto(s)
Proteínas Portadoras , Defectos del Tabique Interventricular , Lisina , Humanos , Animales , Ratones , Linaje de la Célula , Histonas , Acetilación , Cromatina , Factores de Transcripción , Proteínas Supresoras de Tumor , Proteínas de Homeodominio/genética , Proteínas de Ciclo Celular , Histona Acetiltransferasas
3.
Stem Cell Reports ; 19(4): 469-485, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38518784

RESUMEN

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.


Asunto(s)
Neoplasias Hematológicas , Hematopoyesis , Ratones , Animales , Diferenciación Celular/genética , Células Madre Hematopoyéticas , Histona Acetiltransferasas/genética
4.
Cell Rep ; 42(1): 111980, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36641753

RESUMEN

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.


Asunto(s)
Plasticidad de la Célula , Histonas , Histonas/metabolismo , Activación Transcripcional/genética , Acetilación , Plasticidad de la Célula/genética , Células Madre/metabolismo , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo
5.
Cell Death Differ ; 29(5): 1013-1027, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35361962

RESUMEN

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.


Asunto(s)
Factor de Transcripción TFIID , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor , Animales , Apoptosis/genética , Muerte Celular , Ratones , Factor de Transcripción TFIID/genética , Factor de Transcripción TFIID/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
6.
Nature ; 560(7717): 253-257, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30069049

RESUMEN

Acetylation of histones by lysine acetyltransferases (KATs) is essential for chromatin organization and function1. Among the genes coding for the MYST family of KATs (KAT5-KAT8) are the oncogenes KAT6A (also known as MOZ) and KAT6B (also known as MORF and QKF)2,3. KAT6A has essential roles in normal haematopoietic stem cells4-6 and is the target of recurrent chromosomal translocations, causing acute myeloid leukaemia7,8. Similarly, chromosomal translocations in KAT6B have been identified in diverse cancers8. KAT6A suppresses cellular senescence through the regulation of suppressors of the CDKN2A locus9,10, a function that requires its KAT activity10. Loss of one allele of KAT6A extends the median survival of mice with MYC-induced lymphoma from 105 to 413 days11. These findings suggest that inhibition of KAT6A and KAT6B may provide a therapeutic benefit in cancer. Here we present highly potent, selective inhibitors of KAT6A and KAT6B, denoted WM-8014 and WM-1119. Biochemical and structural studies demonstrate that these compounds are reversible competitors of acetyl coenzyme A and inhibit MYST-catalysed histone acetylation. WM-8014 and WM-1119 induce cell cycle exit and cellular senescence without causing DNA damage. Senescence is INK4A/ARF-dependent and is accompanied by changes in gene expression that are typical of loss of KAT6A function. WM-8014 potentiates oncogene-induced senescence in vitro and in a zebrafish model of hepatocellular carcinoma. WM-1119, which has increased bioavailability, arrests the progression of lymphoma in mice. We anticipate that this class of inhibitors will help to accelerate the development of therapeutics that target gene transcription regulated by histone acetylation.


Asunto(s)
Bencenosulfonatos/farmacología , Senescencia Celular/efectos de los fármacos , Histona Acetiltransferasas/antagonistas & inhibidores , Hidrazinas/farmacología , Linfoma/tratamiento farmacológico , Linfoma/patología , Sulfonamidas/farmacología , Acetilación/efectos de los fármacos , Animales , Bencenosulfonatos/uso terapéutico , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Desarrollo de Medicamentos , Fibroblastos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Histona Acetiltransferasas/deficiencia , Histona Acetiltransferasas/genética , Histonas/química , Histonas/metabolismo , Hidrazinas/uso terapéutico , Linfoma/enzimología , Linfoma/genética , Lisina/química , Lisina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Sulfonamidas/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...