Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Curr Protoc ; 3(5): e738, 2023 May.
Article in English | MEDLINE | ID: mdl-37184117

ABSTRACT

Protein lysine acetylation refers to the covalent transfer of an acetyl moiety from acetyl coenzyme A to the epsilon-amino group of a lysine residue and is critical for regulating protein functions in almost all living cells or organisms. Studies in the past decade have demonstrated the unexpected finding that acetylation-like acylation, such as succinylation, propionylation, butyrylation, crotonylation, and lactylation, is also present in histones and many non-histone proteins. Acetylation and acetylation-like acylation serve as reversible on/off switches for regulating protein function while interplaying with other post-translational modifications (such as phosphorylation and methylation) in a codified manner. Lysine acetylation and acetylation-like acylation are important for regulating different cellular and developmental processes in normal and pathological states. Thus, the detection of such modifications is important for related basic research and molecular diagnostics. Traditionally, lysine acetylation is detected by autoradiography, but recent decades have seen great improvement in the quality of site-specific antibodies against acetylation (or acetylation-like acylation), thereby providing competitive alternatives to the use of radioactive acetate and acetyl-coenzyme A for in vivo and in vitro labeling, respectively. This article describes protocols for the detection of lysine acetylation and acetylation-like acylation with site-specific antibodies to complement extant autoradiography-based methods (Pelletier et al., 2017). © 2023 Wiley Periodicals LLC. Basic Protocol 1: Acylation assays in vitro Basic Protocol 2: Determination of in vivo acylation.


Subject(s)
Lysine , Protein Processing, Post-Translational , Acetylation , Lysine/chemistry , Lysine/metabolism , Acylation , Histones/chemistry , Histones/metabolism , Acetyl Coenzyme A/metabolism , Antibodies/metabolism
2.
Am J Hum Genet ; 108(5): 929-941, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33811806

ABSTRACT

Proteins involved in transcriptional regulation harbor a demonstrated enrichment of mutations in neurodevelopmental disorders. The Sin3 (Swi-independent 3)/histone deacetylase (HDAC) complex plays a central role in histone deacetylation and transcriptional repression. Among the two vertebrate paralogs encoding the Sin3 complex, SIN3A variants cause syndromic intellectual disability, but the clinical consequences of SIN3B haploinsufficiency in humans are uncharacterized. Here, we describe a syndrome hallmarked by intellectual disability, developmental delay, and dysmorphic facial features with variably penetrant autism spectrum disorder, congenital malformations, corpus callosum defects, and impaired growth caused by disruptive SIN3B variants. Using chromosomal microarray or exome sequencing, and through international data sharing efforts, we identified nine individuals with heterozygous SIN3B deletion or single-nucleotide variants. Five individuals harbor heterozygous deletions encompassing SIN3B that reside within a ∼230 kb minimal region of overlap on 19p13.11, two individuals have a rare nonsynonymous substitution, and two individuals have a single-nucleotide deletion that results in a frameshift and predicted premature termination codon. To test the relevance of SIN3B impairment to measurable aspects of the human phenotype, we disrupted the orthologous zebrafish locus by genome editing and transient suppression. The mutant and morphant larvae display altered craniofacial patterning, commissural axon defects, and reduced body length supportive of an essential role for Sin3 function in growth and patterning of anterior structures. To investigate further the molecular consequences of SIN3B variants, we quantified genome-wide enhancer and promoter activity states by using H3K27ac ChIP-seq. We show that, similar to SIN3A mutations, SIN3B disruption causes hyperacetylation of a subset of enhancers and promoters in peripheral blood mononuclear cells. Together, these data demonstrate that SIN3B haploinsufficiency leads to a hitherto unknown intellectual disability/autism syndrome, uncover a crucial role of SIN3B in the central nervous system, and define the epigenetic landscape associated with Sin3 complex impairment.


Subject(s)
Autism Spectrum Disorder/genetics , Haploinsufficiency/genetics , Histone Deacetylases/metabolism , Intellectual Disability/genetics , Repressor Proteins/genetics , Acetylation , Adolescent , Animals , Child , Child, Preschool , DNA Copy Number Variations/genetics , Female , Histones/chemistry , Histones/metabolism , Humans , Infant , Larva/genetics , Magnetic Resonance Imaging , Male , Middle Aged , Models, Molecular , Mutation , Repressor Proteins/deficiency , Repressor Proteins/metabolism , Syndrome , Young Adult , Zebrafish/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
3.
HGG Adv ; 2(1): 100015, 2021 Jan 14.
Article in English | MEDLINE | ID: mdl-33537682

ABSTRACT

Histone deacetylases play crucial roles in the regulation of chromatin structure and gene expression in the eukaryotic cell, and disruption of their activity causes a wide range of developmental disorders in humans. Loss-of-function alleles of HDAC4, a founding member of the class IIa deacetylases, have been reported in brachydactyly-mental retardation syndrome (BDMR). However, while disruption of HDAC4 activity and deregulation of its downstream targets may contribute to the BDMR phenotype, loss of HDAC4 function usually occurs as part of larger deletions of chromosome 2q37; BDMR is also known as chromosome 2q37 deletion syndrome, and the precise role of HDAC4 within the phenotype remains uncertain. Thus, identification of missense variants should shed new light on the role of HDAC4 in normal development. Here, we report seven unrelated individuals with a phenotype distinct from that of BDMR, all of whom have heterozygous de novo missense variants that affect a major regulatory site of HDAC4, required for signal-dependent 14-3-3 binding and nucleocytoplasmic shuttling. Two individuals possess variants altering Thr244 or Glu247, whereas the remaining five all carry variants altering Pro248, a key residue for 14-3-3 binding. We propose that the variants in all seven individuals impair 14-3-3 binding (as confirmed for the first two variants by immunoprecipitation assays), thereby identifying deregulation of HDAC4 as a pathological mechanism in a previously uncharacterized developmental disorder.

4.
Am J Hum Genet ; 107(3): 564-574, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32822602

ABSTRACT

KAT5 encodes an essential lysine acetyltransferase, previously called TIP60, which is involved in regulating gene expression, DNA repair, chromatin remodeling, apoptosis, and cell proliferation; but it remains unclear whether variants in this gene cause a genetic disease. Here, we study three individuals with heterozygous de novo missense variants in KAT5 that affect normally invariant residues, with one at the chromodomain (p.Arg53His) and two at or near the acetyl-CoA binding site (p.Cys369Ser and p.Ser413Ala). All three individuals have cerebral malformations, seizures, global developmental delay or intellectual disability, and severe sleep disturbance. Progressive cerebellar atrophy was also noted. Histone acetylation assays with purified variant KAT5 demonstrated that the variants decrease or abolish the ability of the resulting NuA4/TIP60 multi-subunit complexes to acetylate the histone H4 tail in chromatin. Transcriptomic analysis in affected individual fibroblasts showed deregulation of multiple genes that control development. Moreover, there was also upregulated expression of PER1 (a key gene involved in circadian control) in agreement with sleep anomalies in all of the individuals. In conclusion, dominant missense KAT5 variants cause histone acetylation deficiency with transcriptional dysregulation of multiples genes, thereby leading to a neurodevelopmental syndrome with sleep disturbance, cerebellar atrophy, and facial dysmorphisms, and suggesting a recognizable syndrome.


Subject(s)
Atrophy/genetics , Cerebellar Diseases/genetics , Intellectual Disability/genetics , Lysine Acetyltransferase 5/genetics , Abnormalities, Multiple/diagnostic imaging , Abnormalities, Multiple/genetics , Abnormalities, Multiple/physiopathology , Adolescent , Adult , Atrophy/diagnostic imaging , Atrophy/physiopathology , Cerebellar Diseases/diagnostic imaging , Cerebellar Diseases/physiopathology , Child, Preschool , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , DNA Repair/genetics , Epilepsy/diagnostic imaging , Epilepsy/genetics , Epilepsy/physiopathology , Female , Heterozygote , Histones/genetics , Humans , Intellectual Disability/diagnostic imaging , Intellectual Disability/physiopathology , Male , Mutation, Missense/genetics , Protein Processing, Post-Translational/genetics
5.
Sci Adv ; 6(4): eaax0021, 2020 01.
Article in English | MEDLINE | ID: mdl-32010779

ABSTRACT

Lysine acetyltransferase 6A (KAT6A) and its paralog KAT6B form stoichiometric complexes with bromodomain- and PHD finger-containing protein 1 (BRPF1) for acetylation of histone H3 at lysine 23 (H3K23). We report that these complexes also catalyze H3K23 propionylation in vitro and in vivo. Immunofluorescence microscopy and ATAC-See revealed the association of this modification with active chromatin. Brpf1 deletion obliterates the acylation in mouse embryos and fibroblasts. Moreover, we identify BRPF1 variants in 12 previously unidentified cases of syndromic intellectual disability and demonstrate that these cases and known BRPF1 variants impair H3K23 propionylation. Cardiac anomalies are present in a subset of the cases. H3K23 acylation is also impaired by cancer-derived somatic BRPF1 mutations. Valproate, vorinostat, propionate and butyrate promote H3K23 acylation. These results reveal the dual functionality of BRPF1-KAT6 complexes, shed light on mechanisms underlying related developmental disorders and various cancers, and suggest mutation-based therapy for medical conditions with deficient histone acylation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DNA-Binding Proteins/metabolism , Histone Acetyltransferases/metabolism , Histones/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/metabolism , Acetylation , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , Brain/abnormalities , Brain/diagnostic imaging , Cell Line , DNA Mutational Analysis , DNA-Binding Proteins/genetics , Disease Susceptibility , Genetic Predisposition to Disease , Histone Acetyltransferases/genetics , Humans , Magnetic Resonance Imaging , Mice , Mice, Knockout , Models, Biological , Multiprotein Complexes/metabolism , Mutation , Neoplasms/diagnosis , Neurodevelopmental Disorders/diagnosis , Phenotype , Protein Binding , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational , Syndrome
6.
J Clin Invest ; 130(3): 1431-1445, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31794431

ABSTRACT

Epigenetic integrity is critical for many eukaryotic cellular processes. An important question is how different epigenetic regulators control development and influence disease. Lysine acetyltransferase 8 (KAT8) is critical for acetylation of histone H4 at lysine 16 (H4K16), an evolutionarily conserved epigenetic mark. It is unclear what roles KAT8 plays in cerebral development and human disease. Here, we report that cerebrum-specific knockout mice displayed cerebral hypoplasia in the neocortex and hippocampus, along with improper neural stem and progenitor cell (NSPC) development. Mutant cerebrocortical neuroepithelia exhibited faulty proliferation, aberrant neurogenesis, massive apoptosis, and scant H4K16 propionylation. Mutant NSPCs formed poor neurospheres, and pharmacological KAT8 inhibition abolished neurosphere formation. Moreover, we describe KAT8 variants in 9 patients with intellectual disability, seizures, autism, dysmorphisms, and other anomalies. The variants altered chromobarrel and catalytic domains of KAT8, thereby impairing nucleosomal H4K16 acetylation. Valproate was effective for treating epilepsy in at least 2 of the individuals. This study uncovers a critical role of KAT8 in cerebral and NSPC development, identifies 9 individuals with KAT8 variants, and links deficient H4K16 acylation directly to intellectual disability, epilepsy, and other developmental anomalies.


Subject(s)
Hippocampus/enzymology , Histone Acetyltransferases/metabolism , Intellectual Disability/enzymology , Neocortex/enzymology , Neural Stem Cells/enzymology , Acetylation , Animals , HEK293 Cells , Hippocampus/pathology , Histone Acetyltransferases/genetics , Humans , Intellectual Disability/pathology , Mice , Mice, Knockout , Neocortex/pathology , Neural Stem Cells/pathology , Nucleosomes/genetics , Nucleosomes/metabolism
7.
iScience ; 20: 148-167, 2019 Oct 25.
Article in English | MEDLINE | ID: mdl-31569049

ABSTRACT

We report that cerebrum-specific inactivation of the histone deacetylase 3 (HDAC3) gene causes striking developmental defects in the neocortex, hippocampus, and corpus callosum; post-weaning lethality; and abnormal behaviors, including hyperactivity and anxiety. The defects are due to rapid loss of embryonic neural stem and progenitor cells (NSPCs). Premature neurogenesis and abnormal neuronal migration in the mutant brain alter NSPC homeostasis. Mutant cerebral cortices also display augmented DNA damage responses, apoptosis, and histone hyperacetylation. Moreover, mutant NSPCs are impaired in forming neurospheres in vitro, and treatment with the HDAC3-specific inhibitor RGFP966 abolishes neurosphere formation. Transcriptomic analyses of neonatal cerebral cortices and cultured neurospheres support that HDAC3 regulates transcriptional programs through interaction with different transcription factors, including NFIB. These findings establish HDAC3 as a major deacetylase critical for perinatal development of the mouse cerebrum and NSPCs, thereby suggesting a direct link of this enzymatic epigenetic regulator to human cerebral and intellectual development.

8.
Cell Mol Life Sci ; 76(18): 3621-3640, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30953095

ABSTRACT

α-Tubulin acetyltransferase 1 (ATAT1) catalyzes acetylation of α-tubulin at lysine 40 in various organisms ranging from Tetrahymena to humans. Despite the importance in mammals suggested by studies of cultured cells, the mouse Atat1 gene is non-essential for survival, raising an intriguing question about its real functions in vivo. To address this question, we systematically analyzed a mouse strain lacking the gene. The analyses revealed that starting at postnatal day 5, the mutant mice display enlarged lateral ventricles in the forebrain, resembling ventricular dilation in human patients with ventriculomegaly. In the mice, ventricular dilation is due to hypoplasia in the septum and striatum. Behavioral tests of the mice uncovered deficits in motor coordination. Birth-dating experiments revealed that neuronal migration to the mutant septum and striatum is impaired during brain development. In the mutant embryonic fibroblasts, we found mild defects in cell proliferation and primary cilium formation. Notably, in these cells, ATAT1 is indispensable for tubulin hyperacetylation in response to high salt, high glucose, and hydrogen peroxide-induced oxidative stress. We investigated the role of ATAT1 in the hematopoietic system using multicolor flow cytometry and found that this system remains normal in the mutant mice. Although tubulin acetylation was undetectable in a majority of mutant tissues, residual levels were detected in the heart, skeletal muscle, trachea, oviduct, thymus and spleen. This study thus not only establishes the importance of ATAT1 in regulating mouse forebrain development and governing tubulin hyperacetylation during stress responses, but also suggests the existence of an additional α-tubulin acetyltransferase.


Subject(s)
Acetyltransferases/metabolism , Microtubule Proteins/metabolism , Oxidative Stress , Prosencephalon/metabolism , Tubulin/metabolism , Acetylation/drug effects , Acetyltransferases/genetics , Animals , Behavior, Animal , Cell Movement , Cell Proliferation , Cells, Cultured , Cilia/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Hydrogen Peroxide/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule Proteins/genetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , Oxidative Stress/drug effects , Prosencephalon/growth & development , Prosencephalon/pathology
9.
J Biol Chem ; 293(9): 3410-3420, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29321206

ABSTRACT

Short-chain acylation of lysine residues has recently emerged as a group of reversible posttranslational modifications in mammalian cells. The diversity of acylation further broadens the landscape and complexity of the proteome. Identification of regulatory enzymes and effector proteins for lysine acylation is critical to understand functions of these novel modifications at the molecular level. Here, we report that the MYST family of lysine acetyltransferases (KATs) possesses strong propionyltransferase activity both in vitro and in cellulo Particularly, the propionyltransferase activity of MOF, MOZ, and HBO1 is as strong as their acetyltransferase activity. Overexpression of MOF in human embryonic kidney 293T cells induced significantly increased propionylation in multiple histone and non-histone proteins, which shows that the function of MOF goes far beyond its canonical histone H4 lysine 16 acetylation. We also resolved the X-ray co-crystal structure of MOF bound with propionyl-coenzyme A, which provides a direct structural basis for the propionyltransferase activity of the MYST KATs. Our data together define a novel function for the MYST KATs as lysine propionyltransferases and suggest much broader physiological impacts for this family of enzymes.


Subject(s)
Histone Acetyltransferases/metabolism , Protein Processing, Post-Translational , Acetylation , Amino Acid Sequence , HEK293 Cells , Histone Acetyltransferases/chemistry , Humans , Lysine/metabolism , Models, Molecular , Protein Conformation , Proteomics
10.
CRISPR J ; 1: 223-229, 2018 06.
Article in English | MEDLINE | ID: mdl-31021258

ABSTRACT

Precise genome editing using CRISPR typically requires delivery of guide RNAs, Cas9 endonuclease, and DNA repair templates. Both microinjection and electroporation effectively deliver these components into mouse zygotes provided the DNA template is an oligonucleotide of only a few hundred base pairs. However, electroporation completely fails with longer double-stranded DNAs leaving microinjection as the only delivery option. Here, we overcome this limitation by first injecting all CRISPR components, including long plasmid-sized DNA templates, into the sub-zona pellucida space. There they are retained, supporting subsequent electroporation. We show that this simple and well-tolerated method achieves intracellular reagent concentrations sufficient to effect precise gene edits.

11.
Curr Protoc Protein Sci ; 87: 14.11.1-14.11.18, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28150880

ABSTRACT

Lysine acetylation refers to addition of an acetyl moiety to the epsilon-amino group of a lysine residue and is important for regulating protein functions in various organisms from bacteria to humans. This is a reversible and precisely controlled covalent modification that either serves as an on/off switch or participates in a codified manner with other post-translational modifications to regulate different cellular and developmental processes in normal and pathological states. This unit describes methods for in vitro and in vivo determination of lysine acetylation. Such methods can be easily extended for analysis of other acylations (such as propionylation, butyrylation, crotonylation, and succinylation) that are also present in histones and many other proteins. © 2017 by John Wiley & Sons, Inc.


Subject(s)
Lysine/chemistry , Protein Processing, Post-Translational , Proteins/chemistry , Acetylation , Acylation , Animals , Antibodies/immunology , Electrophoresis, Polyacrylamide Gel , HEK293 Cells , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/metabolism , Histones/chemistry , Humans , Lysine Acetyltransferases/chemistry , Lysine Acetyltransferases/metabolism , Rabbits
12.
Am J Hum Genet ; 100(1): 91-104, 2017 Jan 05.
Article in English | MEDLINE | ID: mdl-27939640

ABSTRACT

Identification of over 500 epigenetic regulators in humans raises an interesting question regarding how chromatin dysregulation contributes to different diseases. Bromodomain and PHD finger-containing protein 1 (BRPF1) is a multivalent chromatin regulator possessing three histone-binding domains, one non-specific DNA-binding module, and several motifs for interacting with and activating three lysine acetyltransferases. Genetic analyses of fish brpf1 and mouse Brpf1 have uncovered an important role in skeletal, hematopoietic, and brain development, but it remains unclear how BRPF1 is linked to human development and disease. Here, we describe an intellectual disability disorder in ten individuals with inherited or de novo monoallelic BRPF1 mutations. Symptoms include infantile hypotonia, global developmental delay, intellectual disability, expressive language impairment, and facial dysmorphisms. Central nervous system and spinal abnormalities are also seen in some individuals. These clinical features overlap with but are not identical to those reported for persons with KAT6A or KAT6B mutations, suggesting that BRPF1 targets these two acetyltransferases and additional partners in humans. Functional assays showed that the resulting BRPF1 variants are pathogenic and impair acetylation of histone H3 at lysine 23, an abundant but poorly characterized epigenetic mark. We also found a similar deficiency in different lines of Brpf1-knockout mice. These data indicate that aberrations in the chromatin regulator gene BRPF1 cause histone H3 acetylation deficiency and a previously unrecognized intellectual disability syndrome.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Chromatin/metabolism , Histones/metabolism , Intellectual Disability/genetics , Mutation , Nuclear Proteins/genetics , Acetylation , Adolescent , Alleles , Animals , Carrier Proteins/genetics , Child , Chromatin/chemistry , DNA-Binding Proteins , Developmental Disabilities/genetics , Face/abnormalities , Female , Histone Acetyltransferases/genetics , Humans , Lysine/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Hypotonia/genetics , Syndrome
13.
Proc Natl Acad Sci U S A ; 113(44): 12360-12367, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27791185

ABSTRACT

Translational control of gene expression plays a key role during the early phases of embryonic development. Here we describe a transcriptional regulator of mouse embryonic stem cells (mESCs), Yin-yang 2 (YY2), that is controlled by the translation inhibitors, Eukaryotic initiation factor 4E-binding proteins (4E-BPs). YY2 plays a critical role in regulating mESC functions through control of key pluripotency factors, including Octamer-binding protein 4 (Oct4) and Estrogen-related receptor-ß (Esrrb). Importantly, overexpression of YY2 directs the differentiation of mESCs into cardiovascular lineages. We show that the splicing regulator Polypyrimidine tract-binding protein 1 (PTBP1) promotes the retention of an intron in the 5'-UTR of Yy2 mRNA that confers sensitivity to 4E-BP-mediated translational suppression. Thus, we conclude that YY2 is a major regulator of mESC self-renewal and lineage commitment and document a multilayer regulatory mechanism that controls its expression.


Subject(s)
Alternative Splicing/physiology , Cell Differentiation , Cell Self Renewal/physiology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Transcription Factors/metabolism , Animals , Blastocyst/metabolism , Carrier Proteins/metabolism , Cell Lineage , Cell Self Renewal/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Introns , Mice , Mice, Knockout , Models, Biological , Octamer Transcription Factor-3/metabolism , Phosphoproteins , Polypyrimidine Tract-Binding Protein/genetics , Protein Biosynthesis/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Estrogen/metabolism , Transcription Factors/genetics , Transcription, Genetic/physiology , YY1 Transcription Factor/metabolism
15.
Am J Hum Genet ; 99(4): 934-941, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27616479

ABSTRACT

Chromodomain helicase DNA-binding protein 4 (CHD4) is an ATP-dependent chromatin remodeler involved in epigenetic regulation of gene transcription, DNA repair, and cell cycle progression. Also known as Mi2ß, CHD4 is an integral subunit of a well-characterized histone deacetylase complex. Here we report five individuals with de novo missense substitutions in CHD4 identified through whole-exome sequencing and web-based gene matching. These individuals have overlapping phenotypes including developmental delay, intellectual disability, hearing loss, macrocephaly, distinct facial dysmorphisms, palatal abnormalities, ventriculomegaly, and hypogonadism as well as additional findings such as bone fusions. The variants, c.3380G>A (p.Arg1127Gln), c.3443G>T (p.Trp1148Leu), c.3518G>T (p.Arg1173Leu), and c.3008G>A, (p.Gly1003Asp) (GenBank: NM_001273.3), affect evolutionarily highly conserved residues and are predicted to be deleterious. Previous studies in yeast showed the equivalent Arg1127 and Trp1148 residues to be crucial for SNF2 function. Furthermore, mutations in the same positions were reported in malignant tumors, and a de novo missense substitution in an equivalent arginine residue in the C-terminal helicase domain of SMARCA4 is associated with Coffin Siris syndrome. Cell-based studies of the p.Arg1127Gln and p.Arg1173Leu mutants demonstrate normal localization to the nucleus and HDAC1 interaction. Based on these findings, the mutations potentially alter the complex activity but not its formation. This report provides evidence for the role of CHD4 in human development and expands an increasingly recognized group of Mendelian disorders involving chromatin remodeling and modification.


Subject(s)
Adenosine Triphosphate/metabolism , Autoantigens/genetics , Chromatin Assembly and Disassembly/genetics , Intellectual Disability/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mutation, Missense/genetics , Abnormalities, Multiple/genetics , Adolescent , Animals , Cell Nucleus/metabolism , Child , Child, Preschool , DNA Helicases/genetics , Developmental Disabilities/genetics , Exome/genetics , Face/abnormalities , Female , Hand Deformities, Congenital/genetics , Hearing Loss/genetics , Histone Deacetylase 1/metabolism , Humans , Male , Megalencephaly/genetics , Mice , Micrognathism/genetics , Neck/abnormalities , Nuclear Proteins/genetics , Syndrome , Transcription Factors/genetics
16.
J Clin Invest ; 126(9): 3247-62, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27500495

ABSTRACT

Hematopoietic stem cells (HSCs) serve as a life-long reservoir for all blood cell types and are clinically useful for a variety of HSC transplantation-based therapies. Understanding the role of chromatin organization and regulation in HSC homeostasis may provide important insights into HSC development. Bromodomain- and PHD finger-containing protein 1 (BRPF1) is a multivalent chromatin regulator that possesses 4 nucleosome-binding domains and activates 3 lysine acetyltransferases (KAT6A, KAT6B, and KAT7), suggesting that this protein has the potential to stimulate crosstalk between different chromatin modifications. Here, we investigated the function of BRPF1 in hematopoiesis by selectively deleting its gene in murine blood cells. Brpf1-deficient pups experienced early lethality due to acute bone marrow failure and aplastic anemia. The mutant bone marrow and fetal liver exhibited severe deficiency in HSCs and hematopoietic progenitors, along with elevated reactive oxygen species, senescence, and apoptosis. BRPF1 deficiency also reduced the expression of multipotency genes, including Slamf1, Mecom, Hoxa9, Hlf, Gfi1, Egr, and Gata3. Furthermore, BRPF1 was required for acetylation of histone H3 at lysine 23, a highly abundant but not well-characterized epigenetic mark. These results identify an essential role of the multivalent chromatin regulator BRPF1 in definitive hematopoiesis and illuminate a potentially new avenue for studying epigenetic networks that govern HSC ontogeny.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/physiology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Adaptor Proteins, Signal Transducing , Animals , Apoptosis , Bone Marrow Cells/metabolism , Cellular Senescence , Chromatin/metabolism , DNA-Binding Proteins , Epigenesis, Genetic , Female , Gene Deletion , Hematopoietic Stem Cell Transplantation , Histone Acetyltransferases/metabolism , Histones/metabolism , Homeostasis , Liver/embryology , Male , Mice , Mice, Inbred C57BL , Protein Domains , Reactive Oxygen Species/metabolism , Spleen/metabolism , Thymus Gland/metabolism
17.
Methods Mol Biol ; 1436: 31-45, 2016.
Article in English | MEDLINE | ID: mdl-27246207

ABSTRACT

Histone deacetylases (HDACs) regulate various nuclear and cytoplasmic processes. In mammals, these enzymes are divided into four classes, with class II further divided into two subclasses: IIa (HDAC4, HDAC5, HDAC7, HDAC9) and IIb (HDAC6 and HDAC10). While HDAC6 is mainly cytoplasmic and HDAC10 is pancellular, class IIa HDACs are dynamically shuttled between the nucleus and cytoplasm in a signal-dependent manner, indicating that they are unique signal transducers able to transduce signals from the cytoplasm to chromatin in the nucleus. Once inside the nucleus, class IIa HDACs interact with MEF2 and other transcription factors, mainly acting as transcriptional corepressors. Although class IIa HDACs share many molecular properties in vitro, they play quite distinct roles in vivo. This chapter lists methods that we have used for molecular and biochemical characterization of HDAC4, including development of regular and phospho-specific antibodies, deacetylase activity determination, reporter gene assays, analysis of subcellular localization, and determination of interaction with 14-3-3 and MEF2. Although described specifically for HDAC4, the protocols should be adaptable for analysis to the other three class IIa members, HDAC5, HDAC7, and HDAC9, as well as for other proteins with related properties.


Subject(s)
14-3-3 Proteins/metabolism , Histone Deacetylases/metabolism , MEF2 Transcription Factors/metabolism , Repressor Proteins/metabolism , Animals , Biological Assay , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , Protein Binding
18.
J Biol Chem ; 291(27): 14363-14372, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27143356

ABSTRACT

The adenovirus early region 1A (E1A) oncoprotein hijacks host cells via direct interactions with many key cellular proteins, such as KAT2B, also known as PCAF (p300/CBP associated factor). E1A binds the histone acetyltransferase (HAT) domain of KAT2B to repress its transcriptional activation. However, the molecular mechanism by which E1A inhibits the HAT activity is not known. Here we demonstrate that a short and relatively conserved N-terminal motif (cNM) in the intrinsically disordered E1A protein is crucial for KAT2B interaction, and inhibits its HAT activity through a direct competition with acetyl-CoA, but not its substrate histone H3. Molecular modeling together with a series of mutagenesis experiments suggests that the major helix of E1A cNM binds to a surface of the acetyl-CoA pocket of the KAT2B HAT domain. Moreover, transient expression of the cNM peptide is sufficient to inhibit KAT2B-specific H3 acetylation H3K14ac in vivo Together, our data define an essential motif cNM in N-terminal E1A as an acetyl-CoA entry blocker that directly associates with the entrance of acetyl-CoA binding pocket to block the HAT domain access to its cofactor.


Subject(s)
Adenovirus E1A Proteins/physiology , Lysine Acetyltransferases/antagonists & inhibitors , Acetylation , Adenovirus E1A Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Models, Molecular , Sequence Homology, Amino Acid
19.
EMBO J ; 35(2): 176-92, 2016 Jan 18.
Article in English | MEDLINE | ID: mdl-26620551

ABSTRACT

During DNA replication, thousands of replication origins are activated across the genome. Chromatin architecture contributes to origin specification and usage, yet it remains unclear which chromatin features impact on DNA replication. Here, we perform a RNAi screen for chromatin regulators implicated in replication control by measuring RPA accumulation upon replication stress. We identify six factors required for normal rates of DNA replication and characterize a function of the bromodomain and PHD finger-containing protein 3 (BRPF3) in replication initiation. BRPF3 forms a complex with HBO1 that specifically acetylates histone H3K14, and genomewide analysis shows high enrichment of BRPF3, HBO1 and H3K14ac at ORC1-binding sites and replication origins found in the vicinity of TSSs. Consistent with this, BRPF3 is necessary for H3K14ac at selected origins and efficient origin activation. CDC45 recruitment, but not MCM2-7 loading, is impaired in BRPF3-depleted cells, identifying a BRPF3-dependent function of HBO1 in origin activation that is complementary to its role in licencing. We thus propose that BRPF3-HBO1 acetylation of histone H3K14 around TSS facilitates efficient activation of nearby replication origins.


Subject(s)
Cell Cycle/physiology , Histone Acetyltransferases/metabolism , Histones/metabolism , Replication Origin/physiology , Acetylation , Cell Cycle/genetics , Cell Line , Chromatin/metabolism , Chromatin Immunoprecipitation , DNA Replication/genetics , DNA Replication/physiology , Histone Acetyltransferases/genetics , Humans , Immunohistochemistry , Replication Origin/genetics
20.
J Biol Chem ; 291(6): 2647-63, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26677226

ABSTRACT

To interpret epigenetic information, chromatin readers utilize various protein domains for recognition of DNA and histone modifications. Some readers possess multidomains for modification recognition and are thus multivalent. Bromodomain- and plant homeodomain-linked finger-containing protein 3 (BRPF3) is such a chromatin reader, containing two plant homeodomain-linked fingers, one bromodomain and a PWWP domain. However, its molecular and biological functions remain to be investigated. Here, we report that endogenous BRPF3 preferentially forms a tetrameric complex with HBO1 (also known as KAT7) and two other subunits but not with related acetyltransferases such as MOZ, MORF, TIP60, and MOF (also known as KAT6A, KAT6B, KAT5, and KAT8, respectively). We have also characterized a mutant mouse strain with a lacZ reporter inserted at the Brpf3 locus. Systematic analysis of ß-galactosidase activity revealed dynamic spatiotemporal expression of Brpf3 during mouse embryogenesis and high expression in the adult brain and testis. Brpf3 disruption, however, resulted in no obvious gross phenotypes. This is in stark contrast to Brpf1 and Brpf2, whose loss causes lethality at E9.5 and E15.5, respectively. In Brpf3-null mice and embryonic fibroblasts, RT-quantitative PCR uncovered no changes in levels of Brpf1 and Brpf2 transcripts, confirming no compensation from them. These results indicate that BRPF3 forms a functional tetrameric complex with HBO1 but is not required for mouse development and survival, thereby distinguishing BRPF3 from its paralogs, BRPF1 and BRPF2.


Subject(s)
Embryo, Mammalian/enzymology , Embryonic Development , Histone Acetyltransferases/metabolism , Multienzyme Complexes/metabolism , Animals , Embryo Loss/enzymology , Embryo Loss/genetics , HEK293 Cells , Histone Acetyltransferases/genetics , Humans , Mice , Mice, Mutant Strains , Multienzyme Complexes/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...