Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
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
2.
Hum Mol Genet ; 27(12): 2171-2186, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29648665

ABSTRACT

The human general transcription factor TFIID is composed of the TATA-binding protein (TBP) and 13 TBP-associated factors (TAFs). In eukaryotic cells, TFIID is thought to nucleate RNA polymerase II (Pol II) preinitiation complex formation on all protein coding gene promoters and thus, be crucial for Pol II transcription. In a child with intellectual disability, mild microcephaly, corpus callosum agenesis and poor growth, we identified a homozygous splice-site mutation in TAF8 (NM_138572.2: c.781-1G > A). Our data indicate that the patient's mutation generates a frame shift and an unstable TAF8 mutant protein with an unrelated C-terminus. The mutant TAF8 protein could not be detected in extracts from the patient's fibroblasts, indicating a loss of TAF8 function and that the mutation is most likely causative. Moreover, our immunoprecipitation and proteomic analyses show that in patient cells only partial TAF complexes exist and that the formation of the canonical TFIID is impaired. In contrast, loss of TAF8 in mouse embryonic stem cells and blastocysts leads to cell death and to a global decrease in Pol II transcription. Astonishingly however, in human TAF8 patient cells, we could not detect any cellular phenotype, significant changes in genome-wide Pol II occupancy and pre-mRNA transcription. Thus, the disorganization of the essential holo-TFIID complex did not affect global Pol II transcription in the patient's fibroblasts. Our observations further suggest that partial TAF complexes, and/or an altered TFIID containing a mutated TAF8, could support human development and thus, the absence of holo-TFIID is less deleterious for transcription than originally predicted.


Subject(s)
Intellectual Disability/genetics , Microcephaly/genetics , Transcription Factor TFIID/genetics , Transcription, Genetic , Animals , Blastocyst/metabolism , Cell Death/genetics , Disease Models, Animal , Drosophila/genetics , Homozygote , Humans , Intellectual Disability/diagnostic imaging , Intellectual Disability/physiopathology , Mice , Microcephaly/diagnostic imaging , Microcephaly/pathology , Mouse Embryonic Stem Cells/metabolism , Mutation , RNA Polymerase II/genetics
3.
Proc Natl Acad Sci U S A ; 112(17): 5437-42, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25922517

ABSTRACT

Hox genes underlie the specification of body segment identity in the anterior-posterior axis. They are activated during gastrulation and undergo a dynamic shift from a transcriptionally repressed to an active chromatin state in a sequence that reflects their chromosomal location. Nevertheless, the precise role of chromatin modifying complexes during the initial activation phase remains unclear. In the current study, we examined the role of chromatin regulators during Hox gene activation. Using embryonic stem cell lines lacking the transcriptional activator MOZ and the polycomb-family repressor BMI1, we showed that MOZ and BMI1, respectively, promoted and repressed Hox genes during the shift from the transcriptionally repressed to the active state. Strikingly however, MOZ but not BMI1 was required to regulate Hox mRNA levels after the initial activation phase. To determine the interaction of MOZ and BMI1 in vivo, we interrogated their role in regulating Hox genes and body segment identity using Moz;Bmi1 double deficient mice. We found that the homeotic transformations and shifts in Hox gene expression boundaries observed in single Moz and Bmi1 mutant mice were rescued to a wild type identity in Moz;Bmi1 double knockout animals. Together, our findings establish that MOZ and BMI1 play opposing roles during the onset of Hox gene expression in the ES cell model and during body segment identity specification in vivo. We propose that chromatin-modifying complexes have a previously unappreciated role during the initiation phase of Hox gene expression, which is critical for the correct specification of body segment identity.


Subject(s)
Body Patterning/physiology , Embryo, Mammalian/embryology , Embryonic Stem Cells/metabolism , Histone Acetyltransferases/metabolism , Homeodomain Proteins/biosynthesis , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental/physiology , Histone Acetyltransferases/genetics , Homeodomain Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins/genetics
4.
Stem Cells ; 32(6): 1451-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-25099890

ABSTRACT

Over the past two decades, embryonic stem cells (ESCs) have been established as a valuable system to study the complex molecular events that underlie the collinear activation of Hox genes during development. When ESCs are induced to differentiate in response to retinoic acid (RA), Hox genes are transcriptionally activated in their chromosomal order, with the most 3' Hox genes activated first, sequentially followed by more 5' Hox genes. In contrast to the low levels of RA detected during gastrulation (∼33 nM), a time when Hox genes are induced during embryonic development, high levels of RA are used to study Hox gene activation in ESCs in vitro (1-10 µM). This compelled us to compare RA-induced ESC differentiation in vitro with Hox gene activation in vivo. In this study, we show that treatment of ESCs for 2 days with RA best mimics activation of Hox genes during embryonic development. Furthermore, we show that defects in Hox gene expression known to occur in embryos lacking the histone acetyltransferase MOZ (also called MYST3 or KAT6A) were masked in Moz-deficient ESCs when excessive RA (0.5-5 µM) was used. The role of MOZ in Hox gene activation was only evident when ESCs were differentiated at low concentrations of RA, namely 20 nM, which is similar to RA levels in vivo. Our results demonstrate that using RA at physiologically relevant levels to study the activation of Hox genes, more accurately reflects the molecular events during the early phase of Hox gene activation in vivo.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Tretinoin/pharmacology , Tretinoin/physiology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Embryonic Stem Cells/drug effects , Mice , Mice, Inbred BALB C , Mice, Transgenic
5.
J Med Chem ; 63(9): 4655-4684, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32118427

ABSTRACT

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


Subject(s)
Antineoplastic Agents/pharmacology , Histone Acetyltransferases/antagonists & inhibitors , Hydrazines/pharmacology , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Biological Availability , Drug Discovery , Drug Stability , Humans , Hydrazines/chemical synthesis , Hydrazines/chemistry , Hydrazines/pharmacokinetics , Male , Mice , Microsomes, Liver/metabolism , Molecular Structure , Rats, Sprague-Dawley , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Sulfonamides/metabolism , Sulfonamides/pharmacokinetics
6.
Exp Neurol ; 306: 158-168, 2018 08.
Article in English | MEDLINE | ID: mdl-29772247

ABSTRACT

The distance between nodes of Ranvier, referred to as internode length, positively correlates with axon diameter, and is optimized during development to ensure maximal neuronal conduction velocity. Following myelin loss, internode length is reestablished through remyelination. However, remyelination results in short internode lengths and reduced conduction rates. We analyzed the potential role of neurofilament phosphorylation in regulating internode length during remyelination and myelination. Following ethidium bromide induced demyelination, levels of neurofilament medium (NF-M) and heavy (NF-H) phosphorylation were unaffected. Preventing NF-M lysine-serine-proline (KSP) repeat phosphorylation increased internode length by 30% after remyelination. To further analyze the role of NF-M phosphorylation in regulating internode length, gene replacement was used to produce mice in which all KSP serine residues were replaced with glutamate to mimic constitutive phosphorylation. Mimicking constitutive KSP phosphorylation reduced internode length by 16% during myelination and motor nerve conduction velocity by ~27% without altering sensory nerve structure or function. Our results suggest that NF-M KSP phosphorylation is part of a cooperative mechanism between axons and Schwann cells that together determine internode length, and suggest motor and sensory axons utilize different mechanisms to establish internode length.


Subject(s)
Axons/physiology , Axons/ultrastructure , Motor Neurons/physiology , Motor Neurons/ultrastructure , Myelin Sheath/physiology , Myelin Sheath/ultrastructure , Neurofilament Proteins/metabolism , Remyelination/physiology , Animals , Demyelinating Diseases , Ethidium , Male , Mice , Mutagenesis, Site-Directed , Myelin Sheath/drug effects , Neural Conduction , Neurofilament Proteins/genetics , Phosphorylation , Reaction Time/physiology , Schwann Cells/drug effects , Schwann Cells/ultrastructure , Sciatic Nerve/pathology , Sciatic Nerve/ultrastructure
7.
Ital J Anat Embryol ; 118(1 Suppl): 52-5, 2013.
Article in English | MEDLINE | ID: mdl-24640572

ABSTRACT

The neuropeptide relaxin-3 and its cognate G-protein-coupled receptor, RXFP3, have been implicated in the control of feeding behaviour in rats. For example, relaxin-3-positive projections and RXFP3 are present within hypothalamic feeding circuits, and icv injection of human relaxin-3 (-0.2 to 1.0 nmol) robustly increases feeding behaviour in satiated rats. To explore whether this action is conserved in other experimental species, the present study examined feeding behaviour in C57BL/6J mice following RXFP3 modulation, as mice display near identical regional distribution patterns of relaxin-3/RXFP3, and relaxin-3/RXFP3 signalling has been shown to modulate behavioural arousal in both species. Central injection of the RXFP3 agonists R3/I5 or H3 relaxin (0.5 nmol, icv) did not alter chow consumption in satiated mice relative to vehicle controls, during the 60 min after treatment. Furthermore, relaxin-3 knockout mice displayed similar basal 24-h chow consumption and 1-h palatable food consumption to wildtype littermate controls; although further studies involving acute pharmacological antagonism of RXFP3 in WT mice are required to eliminate the likelihood of compensation in these life-long relaxin-3 deficient mice. Taken together, these findings are in contrast to the potent orexigenic effects of RXFP3 activation observed in rats, and may reflect differential RXFP3 expression within hypothalamic neuron populations in the rat and mouse, or differences in signalling upstream or downstream of relaxin-3/RXFP3 networks in these two species.


Subject(s)
Feeding Behavior/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Neuropeptides/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Relaxin/pharmacology , Animals , Arousal/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orexins , Rats , Relaxin/genetics , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL