Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Nature ; 569(7758): 734-739, 2019 05.
Article in English | MEDLINE | ID: mdl-31118512

ABSTRACT

The execution of developmental programs of gene expression requires an accurate partitioning of the genome into subnuclear compartments, with active euchromatin enriched centrally and silent heterochromatin at the nuclear periphery1. The existence of degenerative diseases linked to lamin A mutations suggests that perinuclear binding of chromatin contributes to cell-type integrity2,3. The methylation of lysine 9 of histone H3 (H3K9me) characterizes heterochromatin and mediates both transcriptional repression and chromatin anchoring at the inner nuclear membrane4. In Caenorhabditis elegans embryos, chromodomain protein CEC-4 bound to the inner nuclear membrane tethers heterochromatin through H3K9me3,5, whereas in differentiated tissues, a second heterochromatin-sequestering pathway is induced. Here we use an RNA interference screen in the cec-4 background and identify MRG-1 as a broadly expressed factor that is necessary for this second chromatin anchor in intestinal cells. However, MRG-1 is exclusively bound to euchromatin, suggesting that it acts indirectly. Heterochromatin detachment in double mrg-1; cec-4 mutants is rescued by depleting the histone acetyltransferase CBP-1/p300 or the transcription factor ATF-8, a member of the bZIP family (which is known to recruit CBP/p300). Overexpression of CBP-1 in cec-4 mutants is sufficient to delocalize heterochromatin in an ATF-8-dependent manner. CBP-1 and H3K27ac levels increase in heterochromatin upon mrg-1 knockdown, coincident with delocalization. This suggests that the spatial organization of chromatin in C. elegans is regulated both by the direct perinuclear attachment of silent chromatin, and by an active retention of CBP-1/p300 in euchromatin. The two pathways contribute differentially in embryos and larval tissues, with CBP-1 sequestration by MRG-1 having a major role in differentiated cells.


Subject(s)
Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Chromatin/genetics , Chromatin/metabolism , Heterochromatin/genetics , Heterochromatin/metabolism , Animals , Caenorhabditis elegans/anatomy & histology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Euchromatin/genetics , Euchromatin/metabolism , Gain of Function Mutation , Genes, Reporter/genetics , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histones/chemistry , Histones/metabolism , Intestines/cytology , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Mol Reprod Dev ; 91(5): e23760, 2024 May.
Article in English | MEDLINE | ID: mdl-38769918

ABSTRACT

e-Lysine acetylation is a prominent histone mark found at transcriptionally active loci. Among many lysine acetyl transferases, nonspecific lethal complex (NSL) members are known to mediate the modification of histone H4. In addition to histone modifications, the KAT8 regulatory complex subunit 3 gene (Kansl3), a core member of NSL complex, has been shown to be involved in several other cellular processes such as mitosis and mitochondrial activity. Although functional studies have been performed on NSL complex members, none of the four core proteins, including Kansl3, have been studied during early mouse development. Here we show that homozygous knockout Kansl3 embryos are lethal at peri-implantation stages, failing to hatch out of the zona pellucida. When the zona pellucida is removed in vitro, Kansl3 null embryos form an abnormal outgrowth with significantly disrupted inner cell mass (ICM) morphology. We document lineage-specific defects at the blastocyst stage with significantly reduced ICM cell number but no difference in trophectoderm cell numbers. Both epiblast and primitive endoderm lineages are altered with reduced cell numbers in null mutants. These results show that Kansl3 is indispensable during early mouse embryonic development and with defects in both ICM and trophectoderm lineages.


Subject(s)
Mice, Knockout , Animals , Mice , Blastocyst Inner Cell Mass/metabolism , Blastocyst Inner Cell Mass/cytology , Female , Embryonic Development , Embryo Loss/pathology , Embryo Loss/genetics , Embryo Loss/metabolism , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Histone Acetyltransferases/deficiency , Blastocyst/metabolism , Blastocyst/cytology
3.
Nature ; 560(7717): 253-257, 2018 08.
Article in English | MEDLINE | ID: mdl-30069049

ABSTRACT

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.


Subject(s)
Benzenesulfonates/pharmacology , Cellular Senescence/drug effects , Histone Acetyltransferases/antagonists & inhibitors , Hydrazines/pharmacology , Lymphoma/drug therapy , Lymphoma/pathology , Sulfonamides/pharmacology , Acetylation/drug effects , Animals , Benzenesulfonates/therapeutic use , Cell Proliferation/drug effects , Cells, Cultured , Drug Development , Fibroblasts , Gene Expression Regulation, Neoplastic/drug effects , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Histones/chemistry , Histones/metabolism , Hydrazines/therapeutic use , Lymphoma/enzymology , Lymphoma/genetics , Lysine/chemistry , Lysine/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Sulfonamides/therapeutic use
4.
J Biol Chem ; 295(25): 8363-8373, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32366460

ABSTRACT

The replisome is a protein complex on the DNA replication fork and functions in a dynamic environment at the intersection of parental and nascent chromatin. Parental nucleosomes are disrupted in front of the replication fork. The daughter DNA duplexes are packaged with an equal amount of parental and newly synthesized histones in the wake of the replication fork through the activity of the replication-coupled chromatin assembly pathway. Histone acetyltransferase 1 (HAT1) is responsible for the cytosolic diacetylation of newly synthesized histone H4 on lysines 5 and 12, which accompanies replication-coupled chromatin assembly. Here, using proximity ligation assay-based chromatin assembly assays and DNA fiber analysis, we analyzed the role of murine HAT1 in replication-coupled chromatin assembly. We demonstrate that HAT1 physically associates with chromatin near DNA replication sites. We found that the association of HAT1 with newly replicated DNA is transient, but can be stabilized by replication fork stalling. The association of HAT1 with nascent chromatin may be functionally relevant, as HAT1 loss decreased replication fork progression and increased replication fork stalling. Moreover, in the absence of HAT1, stalled replication forks were unstable, and newly synthesized DNA became susceptible to MRE11-dependent degradation. These results suggest that HAT1 links replication fork function to the proper processing and assembly of newly synthesized histones.


Subject(s)
DNA Replication , DNA/metabolism , Histone Acetyltransferases/metabolism , Animals , Cell Line , Chromatin/metabolism , Chromatin Assembly and Disassembly , Gene Knockout Techniques , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , MRE11 Homologue Protein/metabolism , Mice
5.
Neurobiol Dis ; 149: 105224, 2021 02.
Article in English | MEDLINE | ID: mdl-33359140

ABSTRACT

The TATA-box binding protein associated factor 1 (TAF1) is part of the TFIID complex that plays a key role during the initiation of transcription. Variants of TAF1 are associated with neurodevelopmental disorders. Previously, we found that CRISPR/Cas9 based editing of the TAF1 gene disrupts the morphology of the cerebral cortex and blunts the expression as well as the function of the CaV3.1 (T-type) voltage gated calcium channel. Here, we tested the efficacy of SAK3 (ethyl 8'-methyl-2', 4-dioxo-2-(piperidin-1-yl)-2'H-spiro [cyclopentane-1, 3'-imidazo [1, 2-a] pyridine]-2-ene-3-carboxylate), a T-type calcium channel enhancer, in an animal model of TAF1 intellectual disability (ID) syndrome. At post-natal day 3, rat pups were subjected to intracerebroventricular (ICV) injection of either gRNA-control or gRNA-TAF1 CRISPR/Cas9 viruses. At post-natal day 21, the rat pups were given SAK3 (0.25 mg/kg, p.o.) or vehicle for 14 days (i.e. till post-natal day 35) and then subjected to behavioral, morphological, and molecular studies. Oral administration of SAK3 (0.25 mg/kg, p.o.) significantly rescued locomotion abnormalities associated with TAF1 gene editing. SAK3 treatment prevented the loss of cortical neurons and GFAP-positive astrocytes observed after TAF1 gene editing. In addition, SAK3 protected cells from apoptosis. SAK3 also restored the Brain-derived neurotrophic factor/protein kinase B/Glycogen Synthase Kinase 3 Beta (BDNF/AKT/GSK3ß) signaling axis in TAF1 edited animals. Finally, SAK3 normalized the levels of three GSK3ß substrates - CaV3.1, FOXP2, and CRMP2. We conclude that the T-type calcium channel enhancer SAK3 is beneficial against the deleterious effects of TAF1 gene-editing, in part, by stimulating the BDNF/AKT/GSK3ß signaling pathway.


Subject(s)
Calcium Channels, T-Type/metabolism , Disease Models, Animal , Histone Acetyltransferases/deficiency , Imidazoles/administration & dosage , Intellectual Disability/drug therapy , Intellectual Disability/metabolism , Spiro Compounds/administration & dosage , TATA-Binding Protein Associated Factors/deficiency , Transcription Factor TFIID/deficiency , Animals , Animals, Newborn , Drug Evaluation, Preclinical/methods , Female , Histone Acetyltransferases/genetics , Injections, Intraventricular , Intellectual Disability/genetics , Locomotion/drug effects , Locomotion/physiology , Pregnancy , Rats , Rats, Sprague-Dawley , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics
6.
Cancer Sci ; 112(5): 1865-1877, 2021 May.
Article in English | MEDLINE | ID: mdl-33544437

ABSTRACT

The histone acetyltransferase MOF (KAT8) is mainly involved in the acetylation of histone H4 at lysine 16 (H4K16) and some non-histone proteins. The MOF expression level is significantly reduced in many cancers, however the biological function of MOF and its underlying mechanism are still elusive in hepatocellular carcinoma (HCC). Estrogen receptor α (ERα) has been considered as a tumor suppressor in HCC. Here, we demonstrated that MOF expression is significantly reduced in HCC samples, and is positively correlated with that of ERα. MOF interacts with ERα, and participates in acetylation of ERα at K266, K268, K299, thereby inhibiting ERα ubiquitination to maintain the stability of ERα. In addition, MOF participates in the upregulation of ERα-mediated transactivation. Depletion of MOF significantly promotes cell growth, migration, and invasion in HCC cell lines. Taken together, our results provide new insights to understand the mechanism underlying the modulation function of MOF on ERα action in HCC, suggesting that MOF might be a potential therapeutic target for HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Estrogen Receptor alpha/metabolism , Histone Acetyltransferases/metabolism , Liver Neoplasms/metabolism , Acetylation , Acetylesterase/metabolism , Animals , Antibodies/therapeutic use , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Movement , Cell Proliferation , Databases, Genetic , Down-Regulation , Estrogen Receptor alpha/genetics , Female , Heterografts , Histone Acetyltransferases/deficiency , Histones/metabolism , Humans , Kaplan-Meier Estimate , Liver Neoplasms/enzymology , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Lysine/metabolism , Male , Mice , Middle Aged , Neoplasm Invasiveness , Signal Transduction , Transcriptional Activation , Ubiquitination , Up-Regulation
7.
Hum Mol Genet ; 28(R2): R254-R264, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31595951

ABSTRACT

The epigenetic machinery in conjunction with the transcriptional machinery is responsible for maintaining genome-wide chromatin states and dynamically regulating gene expression. Mendelian disorders of the epigenetic machinery (MDEMs) are genetic disorders resulting from mutations in components of the epigenetic apparatus. Though individually rare, MDEMs have emerged as a collectively common etiology for intellectual disability (ID) and growth disruption. Studies in model organisms and humans have demonstrated dosage sensitivity of this gene group with haploinsufficiency as a predominant disease mechanism. The epigenetic machinery consists of three enzymatic components (writers, erasers and chromatin remodelers) as well as one non-enzymatic group (readers). A tally of the entire census of such factors revealed that although multiple enzymatic activities never coexist within a single component, individual enzymatic activities often coexist with a reader domain. This group of disorders disrupts both the chromatin and transcription states of target genes downstream of the given component but also DNA methylation on a global scale. Elucidation of these global epigenetic changes may inform our understanding of disease pathogenesis and have diagnostic utility. Moreover, many therapies targeting epigenetic marks already exist, and some have proven successful in treating cancer. This, along with the recent observation that neurological dysfunction in these disorders may in fact be treatable in postnatal life, suggests that the scientific community should prioritize this group as a potentially treatable cause of ID. Here we summarize the recent expansion and major characteristics of MDEMs, as well as the unique therapeutic prospects for this group of disorders.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromatin/enzymology , Epigenesis, Genetic , Intellectual Disability/genetics , Animals , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , DNA Methylation/genetics , Histone Acetyltransferases/deficiency , Humans , Methyltransferases/metabolism , Mice , Rett Syndrome/genetics , Rett Syndrome/metabolism , Sotos Syndrome/enzymology , Sotos Syndrome/genetics , alpha-Thalassemia/genetics
8.
Development ; 144(12): 2165-2174, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28506985

ABSTRACT

Proper oocyte development is crucial for female fertility and requires timely and accurate control of gene expression. K (lysine) acetyltransferase 8 (KAT8), an important component of the X chromosome dosage compensation system in Drosophila, regulates gene activity by acetylating histone H4 preferentially at lysine 16. To explore the function of KAT8 during mouse oocyte development, we crossed Kat8flox/flox mice with Gdf9-Cre mice to specifically delete Kat8 in oocytes. Oocyte Kat8 deletion resulted in female infertility, with follicle development failure in the secondary and preantral follicle stages. RNA-seq analysis revealed that Kat8 deficiency in oocytes results in significant downregulation of antioxidant genes, with a consequent increase in reactive oxygen species. Intraperitoneal injection of the antioxidant N-acetylcysteine rescued defective follicle and oocyte development resulting from Kat8 deficiency. Chromatin immunoprecipitation assays indicated that KAT8 regulates antioxidant gene expression by direct binding to promoter regions. Taken together, our findings demonstrate that KAT8 is essential for female fertility by regulating antioxidant gene expression and identify KAT8 as the first histone acetyltransferase with an essential function in oogenesis.


Subject(s)
Histone Acetyltransferases/metabolism , Oogenesis/physiology , Reactive Oxygen Species/metabolism , Animals , Antioxidants/metabolism , Apoptosis , Female , Fertility/genetics , Fertility/physiology , Gene Expression Regulation, Developmental , Granulosa Cells/metabolism , Heterochromatin/genetics , Heterochromatin/metabolism , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Infertility, Female/genetics , Infertility, Female/metabolism , Infertility, Female/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Oocytes/cytology , Oocytes/metabolism , Oogenesis/genetics , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Pregnancy
9.
J Immunol ; 198(10): 3927-3938, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28424240

ABSTRACT

Histone acetyltransferases (HATs) regulate inducible transcription in multiple cellular processes and during inflammatory and immune response. However, the functions of general control nonrepressed-protein 5 (Gcn5), an evolutionarily conserved HAT from yeast to human, in immune regulation remain unappreciated. In this study, we conditionally deleted Gcn5 (encoded by the Kat2a gene) specifically in T lymphocytes by crossing floxed Gcn5 and Lck-Cre mice, and demonstrated that Gcn5 plays important roles in multiple stages of T cell functions including development, clonal expansion, and differentiation. Loss of Gcn5 functions impaired T cell proliferation, IL-2 production, and Th1/Th17, but not Th2 and regulatory T cell differentiation. Gcn5 is recruited onto the il-2 promoter by interacting with the NFAT in T cells upon TCR stimulation. Interestingly, instead of directly acetylating NFAT, Gcn5 catalyzes histone H3 lysine H9 acetylation to promote IL-2 production. T cell-specific suppression of Gcn5 partially protected mice from myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis, an experimental model for human multiple sclerosis. Our study reveals previously unknown physiological functions for Gcn5 and a molecular mechanism underlying these functions in regulating T cell immunity. Hence Gcn5 may be an important new target for autoimmune disease therapy.


Subject(s)
Histone Acetyltransferases/metabolism , Lymphocyte Activation , T-Lymphocytes/immunology , Animals , Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Gene Expression Regulation , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Interleukin-2/deficiency , Interleukin-2/genetics , Interleukin-2/immunology , Mice , NFATC Transcription Factors/genetics , Promoter Regions, Genetic , Protein Binding , Protein Processing, Post-Translational , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/physiology , Th1 Cells/immunology , Th1 Cells/physiology , Th2 Cells/immunology , Th2 Cells/physiology
10.
Nucleic Acids Res ; 44(15): 7159-72, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27112564

ABSTRACT

The transcriptional response of Saccharomyces cerevisiae to cell wall stress is mainly mediated by the cell wall integrity (CWI) pathway through the MAPK Slt2 and the transcription factor Rlm1. Once activated, Rlm1 interacts with the chromatin remodeling SWI/SNF complex which locally alters nucleosome positioning at the target promoters. Here we show that the SAGA complex plays along with the SWI/SNF complex an important role for eliciting both early induction and sustained gene expression upon stress. Gcn5 co-regulates together with Swi3 the majority of the CWI transcriptional program, except for a group of genes which are only dependent on the SWI/SNF complex. SAGA subunits are recruited to the promoter of CWI-responsive genes in a Slt2, Rlm1 and SWI/SNF-dependent manner. However, Gcn5 mediates acetylation and nucleosome eviction only at the promoters of the SAGA-dependent genes. This process is not essential for pre-initiation transcriptional complex assembly but rather increase the extent of the remodeling mediated by SWI/SNF. As a consequence, H3 eviction and Rlm1 recruitment is completely blocked in a swi3Δ gcn5Δ double mutant. Therefore, SAGA complex, through its histone acetylase activity, cooperates with the SWI/SNF complex for the mandatory nucleosome displacement required for full gene expression through the CWI pathway.


Subject(s)
Gene Expression Regulation, Fungal , Mitogen-Activated Protein Kinases/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Acetylation , Cell Wall/drug effects , Cell Wall/metabolism , Congo Red/toxicity , DNA, Fungal/metabolism , Gene Expression Regulation, Fungal/drug effects , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Histones/metabolism , MADS Domain Proteins/metabolism , Mutation , Promoter Regions, Genetic , Protein Subunits/deficiency , Protein Subunits/genetics , Protein Subunits/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Trans-Activators/chemistry , Trans-Activators/genetics , Transcription, Genetic/drug effects
11.
Nature ; 471(7337): 189-95, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21390126

ABSTRACT

B-cell non-Hodgkin's lymphoma comprises biologically and clinically distinct diseases the pathogenesis of which is associated with genetic lesions affecting oncogenes and tumour-suppressor genes. We report here that the two most common types--follicular lymphoma and diffuse large B-cell lymphoma--harbour frequent structural alterations inactivating CREBBP and, more rarely, EP300, two highly related histone and non-histone acetyltransferases (HATs) that act as transcriptional co-activators in multiple signalling pathways. Overall, about 39% of diffuse large B-cell lymphoma and 41% of follicular lymphoma cases display genomic deletions and/or somatic mutations that remove or inactivate the HAT coding domain of these two genes. These lesions usually affect one allele, suggesting that reduction in HAT dosage is important for lymphomagenesis. We demonstrate specific defects in acetylation-mediated inactivation of the BCL6 oncoprotein and activation of the p53 tumour suppressor. These results identify CREBBP/EP300 mutations as a major pathogenetic mechanism shared by common forms of B-cell non-Hodgkin's lymphoma, with direct implications for the use of drugs targeting acetylation/deacetylation mechanisms.


Subject(s)
Acetyltransferases/genetics , Acetyltransferases/metabolism , CREB-Binding Protein/genetics , E1A-Associated p300 Protein/genetics , Lymphoma, B-Cell/enzymology , Lymphoma, B-Cell/genetics , Mutation/genetics , Acetyl Coenzyme A/metabolism , Acetylation , Acetyltransferases/chemistry , Acetyltransferases/deficiency , Animals , Base Sequence , CREB-Binding Protein/chemistry , CREB-Binding Protein/deficiency , CREB-Binding Protein/metabolism , Cells, Cultured , DNA-Binding Proteins/metabolism , E1A-Associated p300 Protein/chemistry , E1A-Associated p300 Protein/deficiency , E1A-Associated p300 Protein/metabolism , Gene Expression Regulation, Neoplastic , HEK293 Cells , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Humans , Lymphoma, B-Cell/pathology , Lymphoma, Follicular/enzymology , Lymphoma, Follicular/genetics , Lymphoma, Follicular/pathology , Lymphoma, Large B-Cell, Diffuse/enzymology , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mutation, Missense/genetics , Polymorphism, Single Nucleotide/genetics , Protein Binding , Protein Structure, Tertiary/genetics , Proto-Oncogene Proteins c-bcl-6 , Recurrence , Sequence Deletion/genetics , Tumor Suppressor Protein p53/metabolism
12.
Adv Exp Med Biol ; 978: 39-62, 2017.
Article in English | MEDLINE | ID: mdl-28523540

ABSTRACT

Rubinstein-Taybi syndrome (RSTS) is a rare genetic disorder in humans characterized by growth and psychomotor delay, abnormal gross anatomy, and mild to severe mental retardation (Rubinstein and Taybi, Am J Dis Child 105:588-608, 1963, Hennekam et al., Am J Med Genet Suppl 6:56-64, 1990). RSTS is caused by de novo mutations in epigenetics-associated genes, including the cAMP response element-binding protein (CREBBP), the gene-encoding protein referred to as CBP, and the EP300 gene, which encodes the p300 protein, a CBP homologue. Recent studies of the epigenetic mechanisms underlying cognitive functions in mice provide direct evidence for the involvement of nuclear factors (e.g., CBP) in the control of higher cognitive functions. In fact, a role for CBP in higher cognitive function is suggested by the finding that RSTS is caused by heterozygous mutations at the CBP locus (Petrij et al., Nature 376:348-351, 1995). CBP was demonstrated to possess an intrinsic histone acetyltransferase activity (Ogryzko et al., Cell 87:953-959, 1996) that is required for CREB-mediated gene expression (Korzus et al., Science 279:703-707, 1998). The intrinsic protein acetyltransferase activity in CBP might directly destabilize promoter-bound nucleosomes, facilitating the activation of transcription. Due to the complexity of developmental abnormalities and the possible genetic compensation associated with this congenital disorder, however, it is difficult to establish a direct role for CBP in cognitive function in the adult brain. Although aspects of the clinical presentation in RSTS cases have been extensively studied, a spectrum of symptoms found in RSTS patients can be accessed only after birth, and, thus, prenatal genetic tests for this extremely rare genetic disorder are seldom considered. Even though there has been intensive research on the genetic and epigenetic function of the CREBBP gene in rodents, the etiology of this devastating congenital human disorder is largely unknown.


Subject(s)
CREB-Binding Protein/physiology , E1A-Associated p300 Protein/physiology , Epigenesis, Genetic/genetics , Histone Acetyltransferases/physiology , Histone Code/physiology , Nerve Tissue Proteins/physiology , Protein Processing, Post-Translational/genetics , Rubinstein-Taybi Syndrome/genetics , Acetylation , Animals , Brain/metabolism , Brain/pathology , CREB-Binding Protein/deficiency , CREB-Binding Protein/genetics , Cognition/physiology , Disease Models, Animal , E1A-Associated p300 Protein/deficiency , E1A-Associated p300 Protein/genetics , Gene Expression Regulation, Developmental , Genetic Association Studies , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Histone Code/genetics , Histone Deacetylase Inhibitors/therapeutic use , Humans , Invertebrates/genetics , Invertebrates/physiology , Mammals/genetics , Mammals/physiology , Memory/physiology , Models, Neurological , Mutation , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , RNA, Long Noncoding/genetics , Rubinstein-Taybi Syndrome/metabolism
13.
FEMS Yeast Res ; 16(2): fow010, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26851403

ABSTRACT

RTT109 is a histone acetyltransferase for the acetylation of histone H3. It is still not clear whether RTT109 plays a role in regulation of gene expression under environmental stresses. In this study, the involvement of RTT109 in acetic acid stress tolerance of Saccharomyces cerevisiae was investigated. It was revealed that the absence of RTT109 enhanced resistance to 5.5 g L(-1) acetic acid, which was indicated by improved growth of RTT109Δ mutant compared with that of the wild-type BY4741 strain. Meanwhile, the lag phase was shortened for 48 h and glucose consumption completed 36 h in advance for RTT109Δ mutant compared to the wild-type strain, with ethanol production rate increased from 0.39 to 0.60 g L(-1) h(-1). Significantly, elevated transcription levels of HSP12, CTT1 and GSH1, as well as increased activities of antioxidant enzymes were observed in RTT109Δ under acetic acid stress. Improved flocculation of RTT109Δ compared to that of the control strain BY4741 under the acetic acid stress was also observed. These results suggest that the absence of RTT109 not only activates transcription of stress responsive genes, but also improves resistance to oxidative stress, which ultimately contributes to improved acetic acid tolerance in S. cerevisiae.


Subject(s)
Acetic Acid/toxicity , Drug Tolerance , Histone Acetyltransferases/deficiency , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Acetic Acid/metabolism , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation, Fungal , Histone Acetyltransferases/metabolism , Oxidative Stress , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
14.
Proc Natl Acad Sci U S A ; 110(10): 3895-900, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23431171

ABSTRACT

Monocytic leukemia zinc finger (MOZ)/KAT6A is a MOZ, Ybf2/Sas3, Sas2, Tip60 (MYST)-type histone acetyltransferase that functions as a coactivator for acute myeloid leukemia 1 protein (AML1)- and Ets family transcription factor PU.1-dependent transcription. We previously reported that MOZ directly interacts with p53 and is essential for p53-dependent selective regulation of p21 expression. We show here that MOZ is an acetyltransferase of p53 at K120 and K382 and colocalizes with p53 in promyelocytic leukemia (PML) nuclear bodies following cellular stress. The MOZ-PML-p53 interaction enhances MOZ-mediated acetylation of p53, and this ternary complex enhances p53-dependent p21 expression. Moreover, we identified an Akt/protein kinase B recognition sequence in the PML-binding domain of MOZ protein. Akt-mediated phosphorylation of MOZ at T369 has a negative effect on complex formation between PML and MOZ. As a result of PML-mediated suppression of Akt, the increased PML-MOZ interaction enhances p21 expression and induces p53-dependent premature senescence upon forced PML expression. Our research demonstrates that MOZ controls p53 acetylation and transcriptional activity via association with PML.


Subject(s)
Histone Acetyltransferases/metabolism , Leukemia, Promyelocytic, Acute/metabolism , Tumor Suppressor Protein p53/metabolism , Acetylation , Animals , Base Sequence , Cells, Cultured , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Knockout Techniques , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Humans , Intranuclear Inclusion Bodies/metabolism , Leukemia, Promyelocytic, Acute/genetics , Mice , Models, Biological , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tumor Suppressor Protein p53/chemistry , p300-CBP Transcription Factors/metabolism
15.
J Biol Chem ; 289(13): 9365-71, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24554705

ABSTRACT

Quelling and DNA damage-induced small RNA (qiRNA) production are RNA interference (RNAi)-related phenomenon from repetitive genomic loci in Neurospora. We have recently proposed that homologous recombination from repetitive DNA loci allows the RNAi pathway to recognize repetitive DNA to produce small RNA. However, the mechanistic detail of this pathway remains largely unclear. By systematically screening the Neurospora knock-out library, we identified RTT109 as a novel component required for small RNA production. RTT109 is a histone acetyltransferase for histone H3 lysine 56 (H3K56) and H3K56 acetylation is essential for the small RNA biogenesis pathway. Furthermore, we showed that RTT109 is required for homologous recombination and H3K56Ac is enriched around double strand break, which overlaps with RAD51 binding. Taken together, our results suggest that H3K56 acetylation is required for small RNA production through its role in homologous recombination.


Subject(s)
Histones/chemistry , Histones/metabolism , Homologous Recombination/genetics , Lysine/metabolism , RNA Interference , RNA, Small Interfering/biosynthesis , RNA, Small Interfering/genetics , Acetylation , Gene Knockout Techniques , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Neurospora crassa/enzymology , Neurospora crassa/genetics , Neurospora crassa/metabolism
16.
Hum Mol Genet ; 21(6): 1203-16, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22116937

ABSTRACT

Huntington's disease (HD) is an autosomal dominant progressive neurodegenerative disorder caused by an expanded CAG/polyglutamine repeat in the coding region of the huntingtin (htt) gene. Although HD is classically considered a motor disorder, there is now considerable evidence that early cognitive deficits appear in patients before the onset of motor disturbances. Here we demonstrate early impairment of long-term spatial and recognition memory in heterozygous HD knock-in mutant mice (Hdh(Q7/Q111)), a genetically accurate HD mouse model. Cognitive deficits are associated with reduced hippocampal expression of CREB-binding protein (CBP) and diminished levels of histone H3 acetylation. In agreement with reduced CBP, the expression of CREB/CBP target genes related to memory, such c-fos, Arc and Nr4a2, was significantly reduced in the hippocampus of Hdh(Q7/Q111) mice compared with wild-type mice. Finally, and consistent with a role of CBP in cognitive impairment in Hdh(Q7/Q111) mice, administration of the histone deacetylase inhibitor trichostatin A rescues recognition memory deficits and transcription of selective CREB/CBP target genes in Hdh(Q7/Q111) mice. These findings demonstrate an important role for CBP in cognitive dysfunction in HD and suggest the use of histone deacetylase inhibitors as a novel therapeutic strategy for the treatment of memory deficits in this disease.


Subject(s)
CREB-Binding Protein/physiology , Disease Models, Animal , Histone Acetyltransferases/deficiency , Huntington Disease/enzymology , Huntington Disease/pathology , Memory, Long-Term , Acetylation , Animals , Behavior, Animal , Blotting, Western , Cognition Disorders/etiology , Cognition Disorders/pathology , Female , Genes, fos , Hippocampus/metabolism , Hippocampus/pathology , Humans , Immunoenzyme Techniques , Immunoprecipitation , Male , Maze Learning , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
17.
Am J Hum Genet ; 89(5): 675-81, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-22077973

ABSTRACT

Say-Barber-Biesecker-Young-Simpson syndrome (SBBYSS or Ohdo syndrome) is a multiple anomaly syndrome characterized by severe intellectual disability, blepharophimosis, and a mask-like facial appearance. A number of individuals with SBBYSS also have thyroid abnormalities and cleft palate. The condition usually occurs sporadically and is therefore presumed to be due in most cases to new dominant mutations. In individuals with SBBYSS, a whole-exome sequencing approach was used to demonstrate de novo protein-truncating mutations in the highly conserved histone acetyltransferase gene KAT6B (MYST4/MORF)) in three out of four individuals sequenced. Sanger sequencing was used to confirm truncating mutations of KAT6B, clustering in the final exon of the gene in all four individuals and in a further nine persons with typical SBBYSS. Where parental samples were available, the mutations were shown to have occurred de novo. During mammalian development KAT6B is upregulated specifically in the developing central nervous system, facial structures, and limb buds. The phenotypic features seen in the Qkf mouse, a hypomorphic Kat6b mutant, include small eyes, ventrally placed ears and long first digits that mirror the human phenotype. This is a further example of how perturbation of a protein involved in chromatin modification might give rise to a multisystem developmental disorder.


Subject(s)
Codon, Nonsense/genetics , Congenital Hypothyroidism/genetics , Exome/genetics , Histone Acetyltransferases , Intellectual Disability/genetics , Abnormalities, Multiple/genetics , Adult , Animals , Blepharophimosis/genetics , Child , Chromatin/metabolism , Chromosomes, Human, Pair 10/genetics , Facies , Female , Gene Expression Regulation, Developmental , Heart Defects, Congenital , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Humans , INDEL Mutation/genetics , Joint Instability , Male , Metabolism, Inborn Errors/genetics , Mice , Mice, Transgenic , Microarray Analysis , Polymorphism, Single Nucleotide/genetics
18.
J Immunol ; 186(5): 3015-22, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21278346

ABSTRACT

The superoxide anion (O(2)(-))-generating system is an important mechanism of innate immune response against microbial infection in phagocytes and is involved in signal transduction mediated by various physiological and pathological signals in phagocytes and other cells, including B lymphocytes. The O(2)(-)-generating system is composed of five specific proteins: p22-phox, gp91-phox, p40-phox, p47-phox, p67-phox, and a small G protein, Rac. Little is known regarding epigenetic regulation of the genes constituting the O(2)(-)-generating system. In this study, by analyzing the GCN5 (one of most important histone acetyltransferases)-deficient DT40 cell line, we show that GCN5 deficiency causes loss of the O(2)(-)-generating activity. Interestingly, transcription of the gp91-phox gene was drastically downregulated (to ∼4%) in GCN5-deficient cells. To further study the involvement of GCN5 in transcriptional regulation of gp91-phox, we used in vitro differentiation system of U937 cells. When human monoblastic U937 cells were cultured in the presence of IFN-γ, transcription of gp91-phox was remarkably upregulated, and the cells were differentiated to macrophage-like cells that can produce O(2)(-). Chromatin immunoprecipitation assay using the U937 cells during cultivation with IFN-γ revealed not only that association of GCN5 with the gp91-phox gene promoter was significantly accelerated, but also that GCN5 preferentially elevated acetylation levels of H2BK16 and H3K9 surrounding the promoter. These results suggested that GCN5 regulates the O(2)(-)-generating system in leukocytes via controlling the gp91-phox gene expression as a supervisor. Our findings obtained in this study should be useful in understanding the molecular mechanisms involved in epigenetic regulation of the O(2)(-)-generating system in leukocytes.


Subject(s)
Avian Proteins/physiology , Gene Expression Regulation/immunology , Histone Acetyltransferases/physiology , Leukocytes/metabolism , Membrane Glycoproteins/genetics , NADPH Oxidases/genetics , Superoxides/metabolism , p300-CBP Transcription Factors/physiology , Acetylation , Animals , Apoptosis/immunology , Avian Proteins/deficiency , Avian Proteins/genetics , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Line , Chickens , Down-Regulation/immunology , Growth Inhibitors/deficiency , Growth Inhibitors/genetics , Growth Inhibitors/physiology , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Histones/genetics , Histones/metabolism , Humans , Leukocytes/cytology , Leukocytes/enzymology , Lysine/metabolism , Membrane Glycoproteins/biosynthesis , NADPH Oxidases/biosynthesis , Promoter Regions, Genetic/immunology , Superoxides/antagonists & inhibitors , U937 Cells , Up-Regulation/immunology
19.
Proc Natl Acad Sci U S A ; 106(21): 8677-82, 2009 May 26.
Article in English | MEDLINE | ID: mdl-19439667

ABSTRACT

Normal endometrial function requires a balance of progesterone (P4) and estrogen (E2) effects. An imbalance caused by increased E2 action and/or decreased P4 action can result in abnormal endometrial proliferation and, ultimately, endometrial adenocarcinoma, the fourth most common cancer in women. We have identified mitogen-inducible gene 6 (Mig-6) as a downstream target of progesterone receptor (PR) and steroid receptor coactivator (SRC-1) action in the uterus. Here, we demonstrate that absence of Mig-6 in mice results in the inability of P4 to inhibit E2-induced uterine weight gain and E2-responsive target genes expression. At 5 months of age, the absence of Mig-6 results in endometrial hyperplasia. Ovariectomized Mig-6(d/d) mice exhibit this hyperplastic phenotype in the presence of E2 and P4 but not without ovarian hormone. Ovariectomized Mig-6(d/d) mice treated with E2 developed invasive endometrioid-type endometrial adenocarcinoma. Importantly, the observation that endometrial carcinomas from women have a significant reduction in MIG-6 expression provides compelling support for an important growth regulatory role for Mig-6 in the uterus of both humans and mice. This demonstrates the Mig-6 is a critical regulator of the response of the endometrium to E2 in regulating tissue homeostasis. Since Mig-6 is regulated by both PR and SRC-1, this identifies a PR, SRC-1, Mig-6 regulatory pathway that is critical in the suppression of endometrial cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Endometrial Hyperplasia/metabolism , Endometrial Neoplasms/metabolism , Estrogens/metabolism , Progesterone/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adolescent , Adult , Animals , Down-Regulation , Endometrial Hyperplasia/genetics , Endometrial Hyperplasia/pathology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Nuclear Receptor Coactivator 1 , Oligonucleotide Array Sequence Analysis , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins
20.
Mol Cell Biol ; 27(9): 3405-16, 2007 May.
Article in English | MEDLINE | ID: mdl-17325035

ABSTRACT

Gcn5 was the first transcription-related histone acetyltransferase (HAT) to be identified. However, the functions of this enzyme in mammalian cells remain poorly defined. Deletion of Gcn5 in mice leads to early embryonic lethality with increased apoptosis in mesodermal lineages. Here we show that deletion of p53 allows Gcn5(-/-) embryos to survive longer, but Gcn5(-/-) p53(-/-) embryos still die in midgestation. Interestingly, embryos homozygous for point mutations in the Gcn5 catalytic domain survive significantly longer than Gcn5(-/-) or Gcn5(-/-) p53(-/-) mice. In contrast to Gcn5(-/-) embryos, Gcn5(hat/hat) embryos do not exhibit increased apoptosis but do exhibit severe cranial neural tube closure defects and exencephaly. Together, our results indicate that Gcn5 has important, HAT-independent functions in early development and that Gcn5 acetyltransferase activity is required for cranial neural tube closure in the mouse.


Subject(s)
Cell Cycle Proteins/metabolism , Embryo, Mammalian/embryology , Embryo, Mammalian/enzymology , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/metabolism , Neural Tube Defects/enzymology , Neural Tube Defects/pathology , Transcription Factors/deficiency , Transcription Factors/metabolism , Acetylation , Animals , Apoptosis , Biomarkers , Cell Cycle Proteins/genetics , Embryo Loss , Embryo, Mammalian/pathology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Histone Acetyltransferases/genetics , Histones/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Neural Tube Defects/genetics , Neurons/metabolism , Transcription Factors/genetics , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , p300-CBP Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL