Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Nature ; 630(8015): 198-205, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38720074

ABSTRACT

Phosphoinositide-3-kinase-γ (PI3Kγ) is implicated as a target to repolarize tumour-associated macrophages and promote antitumour immune responses in solid cancers1-4. However, cancer cell-intrinsic roles of PI3Kγ are unclear. Here, by integrating unbiased genome-wide CRISPR interference screening with functional analyses across acute leukaemias, we define a selective dependency on the PI3Kγ complex in a high-risk subset that includes myeloid, lymphoid and dendritic lineages. This dependency is characterized by innate inflammatory signalling and activation of phosphoinositide 3-kinase regulatory subunit 5 (PIK3R5), which encodes a regulatory subunit of PI3Kγ5 and stabilizes the active enzymatic complex. We identify p21 (RAC1)-activated kinase 1 (PAK1) as a noncanonical substrate of PI3Kγ that mediates this cell-intrinsic dependency and find that dephosphorylation of PAK1 by PI3Kγ inhibition impairs mitochondrial oxidative phosphorylation. Treatment with the selective PI3Kγ inhibitor eganelisib is effective in leukaemias with activated PIK3R5. In addition, the combination of eganelisib and cytarabine prolongs survival over either agent alone, even in patient-derived leukaemia xenografts with low baseline PIK3R5 expression, as residual leukaemia cells after cytarabine treatment have elevated G protein-coupled purinergic receptor activity and PAK1 phosphorylation. Together, our study reveals a targetable dependency on PI3Kγ-PAK1 signalling that is amenable to near-term evaluation in patients with acute leukaemia.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase , Leukemia , Signal Transduction , p21-Activated Kinases , Animals , Humans , Mice , Cell Line , Class Ib Phosphatidylinositol 3-Kinase/genetics , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Cytarabine/pharmacology , Cytarabine/therapeutic use , Leukemia/drug therapy , Leukemia/enzymology , Leukemia/genetics , Leukemia/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/metabolism , Phosphorylation , Xenograft Model Antitumor Assays
2.
Genes Dev ; 27(18): 2009-24, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-24065767

ABSTRACT

Histone acetyltransferases (HATs) assemble into multisubunit complexes in order to target distinct lysine residues on nucleosomal histones. Here, we characterize native HAT complexes assembled by the BRPF family of scaffold proteins. Their plant homeodomain (PHD)-Zn knuckle-PHD domain is essential for binding chromatin and is restricted to unmethylated H3K4, a specificity that is reversed by the associated ING subunit. Native BRPF1 complexes can contain either MOZ/MORF or HBO1 as catalytic acetyltransferase subunit. Interestingly, while the previously reported HBO1 complexes containing JADE scaffold proteins target histone H4, the HBO1-BRPF1 complex acetylates only H3 in chromatin. We mapped a small region to the N terminus of scaffold proteins responsible for histone tail selection on chromatin. Thus, alternate choice of subunits associated with HBO1 can switch its specificity between H4 and H3 tails. These results uncover a crucial new role for associated proteins within HAT complexes, previously thought to be intrinsic to the catalytic subunit.


Subject(s)
Histone Acetyltransferases/metabolism , Histones/metabolism , Acetylation , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Chromatin/metabolism , DNA-Binding Proteins , HEK293 Cells , HeLa Cells , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/genetics , Homeodomain Proteins/metabolism , Humans , Methylation , Molecular Sequence Data , Nuclear Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Substrate Specificity , Transcription Factors/chemistry , Transcription Factors/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism
3.
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
4.
Nucleic Acids Res ; 44(1): 472-84, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26626149

ABSTRACT

BRPF1 (bromodomain PHD finger 1) is a core subunit of the MOZ histone acetyltransferase (HAT) complex, critical for normal developmental programs and implicated in acute leukemias. BRPF1 contains a unique assembly of zinc fingers, termed a PZP domain, the physiological role of which remains unclear. Here, we elucidate the structure-function relationship of this novel epigenetic reader and detail the biological and mechanistic consequences of its interaction with nucleosomes. PZP has a globular architecture and forms a 2:1 stoichiometry complex with the nucleosome, bivalently interacting with histone H3 and DNA. This binding impacts the nucleosome dynamics, shifting the DNA unwrapping/rewrapping equilibrium toward the unwrapped state and increasing DNA accessibility. We demonstrate that the DNA-binding function of the BRPF1 PZP domain is required for the MOZ-BRPF1-ING5-hEaf6 HAT complex to be recruited to chromatin and to acetylate nucleosomal histones. Our findings reveal a novel link between chromatin dynamics and MOZ-mediated acetylation.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Chromatin/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Protein Interaction Domains and Motifs , Acetylation , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Chromatin/genetics , DNA/chemistry , DNA/metabolism , DNA-Binding Proteins , Histones/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes , Nuclear Proteins/genetics , Nucleosomes/genetics , Protein Binding , Protein Conformation , Protein Stability , Sequence Alignment
5.
PLoS Genet ; 11(3): e1005034, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25757017

ABSTRACT

Lysine acetylation has recently emerged as an important post-translational modification in diverse organisms, but relatively little is known about its roles in mammalian development and stem cells. Bromodomain- and PHD finger-containing protein 1 (BRPF1) is a multidomain histone binder and a master activator of three lysine acetyltransferases, MOZ, MORF and HBO1, which are also known as KAT6A, KAT6B and KAT7, respectively. While the MOZ and MORF genes are rearranged in leukemia, the MORF gene is also mutated in prostate and other cancers and in four genetic disorders with intellectual disability. Here we show that forebrain-specific inactivation of the mouse Brpf1 gene causes hypoplasia in the dentate gyrus, including underdevelopment of the suprapyramidal blade and complete loss of the infrapyramidal blade. We trace the developmental origin to compromised Sox2+ neural stem cells and Tbr2+ intermediate neuronal progenitors. We further demonstrate that Brpf1 loss deregulates neuronal migration, cell cycle progression and transcriptional control, thereby causing abnormal morphogenesis of the hippocampus. These results link histone binding and acetylation control to hippocampus development and identify an important epigenetic regulator for patterning the dentate gyrus, a brain structure critical for learning, memory and adult neurogenesis.


Subject(s)
Carrier Proteins/genetics , Dentate Gyrus/metabolism , Epigenesis, Genetic/genetics , Histone Acetyltransferases/metabolism , Morphogenesis/genetics , Acetylation , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/metabolism , Cell Differentiation/genetics , DNA-Binding Proteins , Dentate Gyrus/growth & development , Dentate Gyrus/pathology , Hippocampus/growth & development , Hippocampus/pathology , Histone Acetyltransferases/genetics , Histones/metabolism , Humans , Mice , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Prosencephalon/embryology , Prosencephalon/growth & development , Prosencephalon/metabolism , Protein Processing, Post-Translational/genetics , T-Box Domain Proteins/genetics
6.
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
7.
J Biol Chem ; 290(18): 11349-64, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25773539

ABSTRACT

With hundreds of chromatin regulators identified in mammals, an emerging issue is how they modulate biological and pathological processes. BRPF1 (bromodomain- and PHD finger-containing protein 1) is a unique chromatin regulator possessing two PHD fingers, one bromodomain and a PWWP domain for recognizing multiple histone modifications. In addition, it binds to the acetyltransferases MOZ, MORF, and HBO1 (also known as KAT6A, KAT6B, and KAT7, respectively) to promote complex formation, restrict substrate specificity, and enhance enzymatic activity. We have recently showed that ablation of the mouse Brpf1 gene causes embryonic lethality at E9.5. Here we present systematic analyses of the mutant animals and demonstrate that the ablation leads to vascular defects in the placenta, yolk sac, and embryo proper, as well as abnormal neural tube closure. At the cellular level, Brpf1 loss inhibits proliferation of embryonic fibroblasts and hematopoietic progenitors. Molecularly, the loss reduces transcription of a ribosomal protein L10 (Rpl10)-like gene and the cell cycle inhibitor p27, and increases expression of the cell-cycle inhibitor p16 and a novel protein homologous to Scp3, a synaptonemal complex protein critical for chromosome association and embryo survival. These results uncover a crucial role of Brpf1 in controlling mouse embryo development and regulating cellular and gene expression programs.


Subject(s)
Carrier Proteins/metabolism , Chromatin/metabolism , Embryonic Development , Adaptor Proteins, Signal Transducing , Animals , Cell Line , Cell Proliferation , DNA-Binding Proteins , Female , Fibroblasts/cytology , Hematopoiesis , Mice , Neovascularization, Physiologic , Neural Tube Defects/metabolism , Placenta/blood supply , Placenta/metabolism , Pregnancy , Yolk Sac/blood supply , Yolk Sac/embryology
8.
J Biol Chem ; 288(18): 12791-804, 2013 May 03.
Article in English | MEDLINE | ID: mdl-23515309

ABSTRACT

Ectopic expression of transcription factors has been shown to reprogram somatic cells into induced pluripotent stem (iPS) cells. It remains largely unexplored how this process is regulated by post-translational modifications. Several reprogramming factors possess conserved sumoylation sites, so we investigated whether and how this modification regulates reprogramming of fibroblasts into iPS cells. Substitution of the sole sumoylation site of the Krüppel-like factor (KLF4), a well known reprogramming factor, promoted iPS cell formation. In comparison, much smaller effects on reprogramming were observed for sumoylation-deficient mutants of SOX2 and OCT4, two other classical reprogramming factors. We also analyzed KLF2, a KLF4 homolog and a member of the KLF family of transcription factors with a known role in reprogramming. KLF2 was sumoylated at two conserved neighboring motifs, but substitution of the key lysine residues only stimulated reprogramming slightly. KLF5 is another KLF member with an established link to embryonic stem cell pluripotency. Interestingly, although it was much more efficiently sumoylated than either KLF2 or KLF4, KLF5 was inactive in reprogramming, and its sumoylation was not responsible for this deficiency. Furthermore, sumoylation of KLF4 but not KLF2 or KLF5 stimulated adipocyte differentiation. These results thus demonstrate the importance KLF4 sumoylation in regulating pluripotency and adipocyte differentiation.


Subject(s)
Adipocytes/metabolism , Cell Differentiation , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Transcription Factors/metabolism , Sumoylation , 3T3-L1 Cells , Adipocytes/cytology , Animals , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
9.
J Biol Chem ; 288(8): 5591-605, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23297420

ABSTRACT

Histone deacetylase 4 (HDAC4) and its paralogs, HDAC5, -7, and -9 (all members of class IIa), possess multiple phosphorylation sites crucial for 14-3-3 binding and subsequent nuclear export. cAMP signaling stimulates nuclear import of HDAC4 and HDAC5, but the underlying mechanisms remain to be elucidated. Here we show that cAMP potentiates nuclear localization of HDAC9. Mutation of an SP motif conserved in HDAC4, -5, and -9 prevents cAMP-stimulated nuclear localization. Unexpectedly, this treatment inhibits phosphorylation at the SP motif, indicating an inverse relationship between the phosphorylation event and nuclear import. Consistent with this, leptomycin B-induced nuclear import and adrenocorticotropic hormone (ACTH) treatment result in the dephosphorylation at the motif. Moreover, the modification synergizes with phosphorylation at a nearby site, and similar kinetics was observed for both phosphorylation events during myoblast and adipocyte differentiation. These results thus unravel a previously unrecognized mechanism whereby cAMP promotes dephosphorylation and differentially regulates multisite phosphorylation and the nuclear localization of class IIa HDACs.


Subject(s)
Cyclic AMP/metabolism , Histone Deacetylases/biosynthesis , 3T3 Cells , Active Transport, Cell Nucleus , Amino Acid Motifs , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoplasm/metabolism , HEK293 Cells , HeLa Cells , Histone Deacetylases/chemistry , Humans , Insecta , Mice , Phosphorylation , Plasmids/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL