Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
J Clin Invest ; 134(4)2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38175723

ABSTRACT

Aster proteins mediate the nonvesicular transport of cholesterol from the plasma membrane (PM) to the endoplasmic reticulum (ER). However, the importance of nonvesicular sterol movement for physiology and pathophysiology in various tissues is incompletely understood. Here we show that loss of Aster-B leads to diet-induced obesity in female but not in male mice, and that this sex difference is abolished by ovariectomy. We further demonstrate that Aster-B deficiency impairs nonvesicular cholesterol transport from the PM to the ER in ovaries in vivo, leading to hypogonadism and reduced estradiol synthesis. Female Aster-B-deficient mice exhibit reduced locomotor activity and energy expenditure, consistent with established effects of estrogens on systemic metabolism. Administration of exogenous estradiol ameliorates the diet-induced obesity phenotype of Aster-B-deficient female mice. These findings highlight the key role of Aster-B-dependent nonvesicular cholesterol transport in regulating estradiol production and protecting females from obesity.


Subject(s)
Cholesterol , Estradiol , Female , Mice , Male , Animals , Estradiol/metabolism , Cell Membrane/metabolism , Cholesterol/metabolism , Obesity/genetics , Obesity/metabolism , Diet
2.
Life Sci Alliance ; 7(1)2024 01.
Article in English | MEDLINE | ID: mdl-37833074

ABSTRACT

About a quarter of total human cancers carry mutations in Ras isoforms. Accumulating evidence suggests that small GTPases, RalA, and RalB, and their activators, Ral guanine nucleotide exchange factors (RalGEFs), play an essential role in oncogenic Ras-induced signalling. We studied the interaction between human KRas4B and the Ras association (RA) domain of Rgl2 (Rgl2RA), one of the RA-containing RalGEFs. We show that the G12V oncogenic KRas4B mutation changes the interaction kinetics with Rgl2RA The crystal structure of the KRas4BG12V: Rgl2RA complex shows a 2:2 heterotetramer where the switch I and switch II regions of each KRasG12V interact with both Rgl2RA molecules. This structural arrangement is highly similar to the HRasE31K:RALGDSRA crystal structure and is distinct from the well-characterised Ras:Raf complex. Interestingly, the G12V mutation was found at the dimer interface of KRas4BG12V with its partner. Our study reveals a potentially distinct mode of Ras:effector complex formation by RalGEFs and offers a possible mechanistic explanation for how the oncogenic KRas4BG12V hyperactivates the RalA/B pathway.


Subject(s)
Monomeric GTP-Binding Proteins , Humans , Monomeric GTP-Binding Proteins/metabolism , Signal Transduction/genetics , Protein Isoforms/metabolism , Genes, ras
3.
Science ; 382(6671): eadf0966, 2023 11 10.
Article in English | MEDLINE | ID: mdl-37943936

ABSTRACT

Intestinal absorption is an important contributor to systemic cholesterol homeostasis. Niemann-Pick C1 Like 1 (NPC1L1) assists in the initial step of dietary cholesterol uptake, but how cholesterol moves downstream of NPC1L1 is unknown. We show that Aster-B and Aster-C are critical for nonvesicular cholesterol movement in enterocytes. Loss of NPC1L1 diminishes accessible plasma membrane (PM) cholesterol and abolishes Aster recruitment to the intestinal brush border. Enterocytes lacking Asters accumulate PM cholesterol and show endoplasmic reticulum cholesterol depletion. Aster-deficient mice have impaired cholesterol absorption and are protected against diet-induced hypercholesterolemia. Finally, the Aster pathway can be targeted with a small-molecule inhibitor to manipulate cholesterol uptake. These findings identify the Aster pathway as a physiologically important and pharmacologically tractable node in dietary lipid absorption.


Subject(s)
Cholesterol, Dietary , Enterocytes , Intestinal Absorption , Membrane Transport Proteins , Animals , Mice , Biological Transport , Cholesterol, Dietary/metabolism , Intestinal Absorption/drug effects , Intestinal Absorption/physiology , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice, Inbred C57BL , Enterocytes/metabolism , Liver X Receptors/metabolism , Humans , Jejunum/metabolism , Mice, Knockout
4.
bioRxiv ; 2023 Jul 10.
Article in English | MEDLINE | ID: mdl-37503112

ABSTRACT

Intestinal cholesterol absorption is an important contributor to systemic cholesterol homeostasis. Niemann-Pick C1 Like 1 (NPC1L1), the target of the drug ezetimibe (EZ), assists in the initial step of dietary cholesterol uptake. However, how cholesterol moves downstream of NPC1L1 is unknown. Here we show that Aster-B and Aster-C are critical for non-vesicular cholesterol movement in enterocytes, bridging NPC1L1 at the plasma membrane (PM) and ACAT2 in the endoplasmic reticulum (ER). Loss of NPC1L1 diminishes accessible PM cholesterol in enterocytes and abolishes Aster recruitment to the intestinal brush border. Enterocytes lacking Asters accumulate cholesterol at the PM and display evidence of ER cholesterol depletion, including decreased cholesterol ester stores and activation of the SREBP-2 transcriptional pathway. Aster-deficient mice have impaired cholesterol absorption and are protected against diet-induced hypercholesterolemia. Finally, we show that the Aster pathway can be targeted with a small molecule inhibitor to manipulate dietary cholesterol uptake. These findings identify the Aster pathway as a physiologically important and pharmacologically tractable node in dietary lipid absorption. One-Sentence Summary: Identification of a targetable pathway for regulation of dietary cholesterol absorption.

5.
Nucleic Acids Res ; 51(12): 6006-6019, 2023 07 07.
Article in English | MEDLINE | ID: mdl-37099381

ABSTRACT

Histone deacetylases 1 and 2 (HDAC1/2) serve as the catalytic subunit of six distinct families of nuclear complexes. These complexes repress gene transcription through removing acetyl groups from lysine residues in histone tails. In addition to the deacetylase subunit, these complexes typically contain transcription factor and/or chromatin binding activities. The MIER:HDAC complex has hitherto been poorly characterized. Here, we show that MIER1 unexpectedly co-purifies with an H2A:H2B histone dimer. We show that MIER1 is also able to bind a complete histone octamer. Intriguingly, we found that a larger MIER1:HDAC1:BAHD1:C1QBP complex additionally co-purifies with an intact nucleosome on which H3K27 is either di- or tri-methylated. Together this suggests that the MIER1 complex acts downstream of PRC2 to expand regions of repressed chromatin and could potentially deposit histone octamer onto nucleosome-depleted regions of DNA.


Subject(s)
Histone Deacetylases , Nucleosomes , Chromatin/genetics , Histone Deacetylases/metabolism , Histones/metabolism , Multiprotein Complexes/metabolism , Nucleosomes/genetics , Transcription Factors/metabolism , Humans
6.
ACS Chem Biol ; 17(9): 2572-2582, 2022 09 16.
Article in English | MEDLINE | ID: mdl-35973051

ABSTRACT

Targeting the lysine deacetylase activity of class I histone deacetylases (HDACs) is potentially beneficial for the treatment of several diseases including human immunodeficiency virus (HIV) infection, Alzheimer's disease, and various cancers. It is therefore important to understand the function and mechanism of action of these enzymes. Class I HDACs act as catalytic components of seven large, multiprotein corepressor complexes. Different HDAC corepressor complexes have specific, nonredundant roles in the cell. It is likely that their specific functions are at least partly influenced by the substrate specificity of the complexes. To address this, we developed chemical tools to probe the specificity of HDAC complexes. We assessed a library of acetyl-lysine-containing substrate peptides and hydroxamic acid-containing inhibitor peptides against the full range of class I HDAC corepressor complexes. The results suggest that site-specific HDAC corepressor complex activity is driven in part by the recognition of the primary amino acid sequence surrounding a particular lysine position in the histone tail.


Subject(s)
Hydroxamic Acids , Peptide Library , Co-Repressor Proteins/metabolism , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Hydroxamic Acids/chemistry , Lysine , Peptides/chemistry
7.
PLoS Pathog ; 18(7): e1010733, 2022 07.
Article in English | MEDLINE | ID: mdl-35849637

ABSTRACT

Emerging SARS-CoV-2 variants are creating major challenges in the ongoing COVID-19 pandemic. Being able to predict mutations that could arise in SARS-CoV-2 leading to increased transmissibility or immune evasion would be extremely valuable in development of broad-acting therapeutics and vaccines, and prioritising viral monitoring and containment. Here we use in vitro evolution to seek mutations in SARS-CoV-2 receptor binding domain (RBD) that would substantially increase binding to ACE2. We find a double mutation, S477N and Q498H, that increases affinity of RBD for ACE2 by 6.5-fold. This affinity gain is largely driven by the Q498H mutation. We determine the structure of the mutant-RBD:ACE2 complex by cryo-electron microscopy to reveal the mechanism for increased affinity. Addition of Q498H to SARS-CoV-2 RBD variants is found to boost binding affinity of the variants for human ACE2 and confer a new ability to bind rat ACE2 with high affinity. Surprisingly however, in the presence of the common N501Y mutation, Q498H inhibits binding, due to a clash between H498 and Y501 side chains. To achieve an intermolecular bonding network, affinity gain and cross-species binding similar to Q498H alone, RBD variants with the N501Y mutation must acquire instead the related Q498R mutation. Thus, SARS-CoV-2 RBD can access large affinity gains and cross-species binding via two alternative mutational routes involving Q498, with route selection determined by whether a variant already has the N501Y mutation. These mutations are now appearing in emerging SARS-CoV-2 variants where they have the potential to influence human-to-human and cross-species transmission.


Subject(s)
COVID-19 , SARS-CoV-2 , Angiotensin-Converting Enzyme 2/genetics , Animals , COVID-19/genetics , Cryoelectron Microscopy , Humans , Mutation , Pandemics , Peptidyl-Dipeptidase A/metabolism , Protein Binding , Rats , Receptors, Virus/metabolism , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/metabolism
8.
J Med Chem ; 65(7): 5642-5659, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35293758

ABSTRACT

Class I histone deacetylase (HDAC) enzymes 1, 2, and 3 organize chromatin as the catalytic subunits within seven distinct multiprotein corepressor complexes and are established drug targets. We report optimization studies of benzamide-based Von Hippel-Lindau (VHL) E3-ligase proteolysis targeting chimeras (PROTACs) and for the first time describe transcriptome perturbations resulting from these degraders. By modifying the linker and VHL ligand, we identified PROTACs 7, 9, and 22 with submicromolar DC50 values for HDAC1 and/or HDAC3 in HCT116 cells. A hook effect was observed for HDAC3 that could be negated by modifying the position of attachment of the VHL ligand to the linker. The more potent HDAC1/2 degraders correlated with greater total differentially expressed genes and enhanced apoptosis in HCT116 cells. We demonstrate that HDAC1/2 degradation by PROTACs correlates with enhanced global gene expression and apoptosis, important for the development of more efficacious HDAC therapeutics with reduced side effects.


Subject(s)
Histone Deacetylases , Neoplasms , Apoptosis , Chimera/metabolism , Histone Deacetylase 1/metabolism , Histone Deacetylases/metabolism , Humans , Ligands , Neoplasms/drug therapy , Proteolysis , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
9.
J Am Chem Soc ; 144(8): 3360-3364, 2022 03 02.
Article in English | MEDLINE | ID: mdl-35175758

ABSTRACT

We describe a new method to produce histone H2B by semisynthesis with an engineered sortase transpeptidase. N-Terminal tail site-specifically modified acetylated, lactylated, and ß-hydroxybutyrylated histone H2Bs were incorporated into nucleosomes and investigated as substrates of histone deacetylase (HDAC) complexes and sirtuins. A wide range of rates and site-specificities were observed by these enzyme forms suggesting distinct biological roles in regulating chromatin structure and epigenetics.


Subject(s)
Histones , Sirtuins , Chromatin , Histone Deacetylases/genetics , Histones/chemistry , Nucleosomes
10.
Mol Cell Biol ; 42(2): e0036321, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34871063

ABSTRACT

Mutations in thyroid hormone receptor α (TRα), a ligand-inducible transcription factor, cause resistance to thyroid hormone α (RTHα). This disorder is characterized by tissue-specific hormone refractoriness and hypothyroidism due to the inhibition of target gene expression by mutant TRα-corepressor complexes. Using biophysical approaches, we show that RTHα-associated TRα mutants devoid of ligand-dependent transcription activation function unexpectedly retain the ability to bind thyroid hormone. Visualization of the ligand T3 within the crystal structure of a prototypic TRα mutant validates this notion. This finding prompted the synthesis of different thyroid hormone analogues, identifying a lead compound, ES08, which dissociates corepressor from mutant human TRα more efficaciously than T3. ES08 rescues developmental anomalies in a zebrafish model of RTHα and induces target gene expression in TRα mutation-containing cells from an RTHα patient more effectively than T3. Our observations provide proof of principle for developing synthetic ligands that can relieve transcriptional repression by the mutant TRα-corepressor complex for treatment of RTHα.


Subject(s)
Co-Repressor Proteins/genetics , Gene Expression/physiology , Genetic Predisposition to Disease/genetics , Thyroid Hormones/metabolism , Animals , Humans , Mutation/genetics , Phenotype , Receptors, Thyroid Hormone/genetics , Thyroid Hormone Receptors alpha/metabolism , Triiodothyronine/genetics
11.
Nat Commun ; 12(1): 819, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33547302

ABSTRACT

Regulated cell death is essential in development and cellular homeostasis. Multi-protein platforms, including the Death-Inducing Signaling Complex (DISC), co-ordinate cell fate via a core FADD:Caspase-8 complex and its regulatory partners, such as the cell death inhibitor c-FLIP. Here, using electron microscopy, we visualize full-length procaspase-8 in complex with FADD. Our structural analysis now reveals how the FADD-nucleated tandem death effector domain (tDED) helical filament is required to orientate the procaspase-8 catalytic domains, enabling their activation via anti-parallel dimerization. Strikingly, recruitment of c-FLIPS into this complex inhibits Caspase-8 activity by altering tDED triple helix architecture, resulting in steric hindrance of the canonical tDED Type I binding site. This prevents both Caspase-8 catalytic domain assembly and tDED helical filament elongation. Our findings reveal how the plasticity, composition and architecture of the core FADD:Caspase-8 complex critically defines life/death decisions not only via the DISC, but across multiple key signaling platforms including TNF complex II, the ripoptosome, and RIPK1/RIPK3 necrosome.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/chemistry , Caspase 8/chemistry , Fas-Associated Death Domain Protein/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Catalytic Domain , Cloning, Molecular , Cryoelectron Microscopy , Death Domain Receptor Signaling Adaptor Proteins/chemistry , Death Domain Receptor Signaling Adaptor Proteins/genetics , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Multimerization , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Regulated Cell Death/genetics , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
12.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article in English | MEDLINE | ID: mdl-33376205

ABSTRACT

The Aster proteins (encoded by the Gramd1a-c genes) contain a ligand-binding fold structurally similar to a START domain and mediate nonvesicular plasma membrane (PM) to endoplasmic reticulum (ER) cholesterol transport. In an effort to develop small molecule modulators of Asters, we identified 20α-hydroxycholesterol (HC) and U18666A as lead compounds. Unfortunately, both 20α-HC and U18666A target other sterol homeostatic proteins, limiting their utility. 20α-HC inhibits sterol regulatory element-binding protein 2 (SREBP2) processing, and U18666A is an inhibitor of the vesicular trafficking protein Niemann-Pick C1 (NPC1). To develop potent and selective Aster inhibitors, we synthesized a series of compounds by modifying 20α-HC and U18666A. Among these, AI (Aster inhibitor)-1l, which has a longer side chain than 20α-HC, selectively bound to Aster-C. The crystal structure of Aster-C in complex with AI-1l suggests that sequence and flexibility differences in the loop that gates the binding cavity may account for the ligand specificity for Aster C. We further identified the U18666A analog AI-3d as a potent inhibitor of all three Aster proteins. AI-3d blocks the ability of Asters to bind and transfer cholesterol in vitro and in cells. Importantly, AI-3d also inhibits the movement of low-density lipoprotein (LDL) cholesterol to the ER, although AI-3d does not block NPC1. This finding positions the nonvesicular Aster pathway downstream of NPC1-dependent vesicular transport in the movement of LDL cholesterol to the ER. Selective Aster inhibitors represent useful chemical tools to distinguish vesicular and nonvesicular sterol transport mechanisms in mammalian cells.


Subject(s)
Biological Transport/drug effects , Membrane Glycoproteins/drug effects , Membrane Proteins/metabolism , Androstenes/pharmacology , Animals , CHO Cells , Carrier Proteins/metabolism , Cell Membrane/metabolism , Cholesterol/metabolism , Cholesterol, LDL/metabolism , Cricetulus , Endoplasmic Reticulum/metabolism , Humans , Hydroxycholesterols/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/physiology , Niemann-Pick C1 Protein/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism , Sterols/metabolism
13.
Nucleic Acids Res ; 48(22): 12972-12982, 2020 12 16.
Article in English | MEDLINE | ID: mdl-33264408

ABSTRACT

Class I histone deacetylase complexes play essential roles in many nuclear processes. Whilst they contain a common catalytic subunit, they have diverse modes of action determined by associated factors in the distinct complexes. The deacetylase module from the NuRD complex contains three protein domains that control the recruitment of chromatin to the deacetylase enzyme, HDAC1/2. Using biochemical approaches and cryo-electron microscopy, we have determined how three chromatin-binding domains (MTA1-BAH, MBD2/3 and RBBP4/7) are assembled in relation to the core complex so as to facilitate interaction of the complex with the genome. We observe a striking arrangement of the BAH domains suggesting a potential mechanism for binding to di-nucleosomes. We also find that the WD40 domains from RBBP4 are linked to the core with surprising flexibility that is likely important for chromatin engagement. A single MBD2 protein binds asymmetrically to the dimerisation interface of the complex. This symmetry mismatch explains the stoichiometry of the complex. Finally, our structures suggest how the holo-NuRD might assemble on a di-nucleosome substrate.


Subject(s)
Chromatin/genetics , DNA-Binding Proteins/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Repressor Proteins/genetics , Retinoblastoma-Binding Protein 4/genetics , Trans-Activators/genetics , Amino Acid Sequence/genetics , Cryoelectron Microscopy , DNA-Binding Proteins/ultrastructure , Histone Deacetylase 1/genetics , Histone Deacetylase 1/ultrastructure , Histone Deacetylases/genetics , Histone Deacetylases/ultrastructure , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/ultrastructure , Nucleosomes/genetics , Nucleosomes/ultrastructure , Protein Binding/genetics , Protein Domains/genetics , Repressor Proteins/ultrastructure , Retinoblastoma-Binding Protein 4/ultrastructure , Trans-Activators/ultrastructure
14.
Proc Natl Acad Sci U S A ; 117(38): 23597-23605, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32900932

ABSTRACT

Trinucleotide repeat (TNR) expansions cause nearly 20 severe human neurological diseases which are currently untreatable. For some of these diseases, ongoing somatic expansions accelerate disease progression and may influence age of onset. This new knowledge emphasizes the importance of understanding the protein factors that drive expansions. Recent genetic evidence indicates that the mismatch repair factor MutSß (Msh2-Msh3 complex) and the histone deacetylase HDAC3 function in the same pathway to drive triplet repeat expansions. Here we tested the hypothesis that HDAC3 deacetylates MutSß and thereby activates it to drive expansions. The HDAC3-selective inhibitor RGFP966 was used to examine its biological and biochemical consequences in human tissue culture cells. HDAC3 inhibition efficiently suppresses repeat expansion without impeding canonical mismatch repair activity. Five key lysine residues in Msh3 are direct targets of HDAC3 deacetylation. In cells expressing Msh3 in which these lysine residues are mutated to arginine, the inhibitory effect of RGFP966 on expansions is largely bypassed, consistent with the direct deacetylation hypothesis. RGFP966 treatment does not alter MutSß subunit abundance or complex formation but does partially control its subcellular localization. Deacetylation sites in Msh3 overlap a nuclear localization signal, and we show that localization of MutSß is partially dependent on HDAC3 activity. Together, these results indicate that MutSß is a key target of HDAC3 deacetylation and provide insights into an innovative regulatory mechanism for triplet repeat expansions. The results suggest expansion activity may be druggable and support HDAC3-selective inhibition as an attractive therapy in some triplet repeat expansion diseases.


Subject(s)
DNA Mismatch Repair/genetics , Histone Deacetylases , Trinucleotide Repeat Expansion/genetics , Acetylation/drug effects , Acrylamides/pharmacology , Cell Line , Cells, Cultured , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Phenylenediamines/pharmacology
15.
Thyroid ; 30(11): 1681-1684, 2020 11.
Article in English | MEDLINE | ID: mdl-32669045

ABSTRACT

A 23-year-old man and his grandmother with hyperthyroxinemia and hypercortisolemia were heterozygous for an ALB mutation (p. Arg218Pro), known to cause familial dysalbuminemic hyperthyroxinemia (FDH). However, serum-free cortisol levels in these individuals were normal and total cortisol concentrations fell markedly after depletion of albumin from their serum. We conclude that binding of steroid as well as iodothyronines to mutant albumin causes raised circulating cortisol as well as thyroid hormones in euthyroid euadrenal individuals with R218P FDH, with potential for misdiagnosis, unnecessary investigation, and inappropriate treatment.


Subject(s)
Hydrocortisone/blood , Hyperthyroxinemia, Familial Dysalbuminemic/complications , Hyperthyroxinemia/complications , Mutation , Serum Albumin, Human/genetics , Albumins/chemistry , Genotype , Heterozygote , Humans , Immunoassay , Male , Military Personnel , Protein Binding , Serum Albumin/genetics , Steroids/chemistry , Thyronines/blood , Thyroxine/blood , Young Adult
16.
Nat Commun ; 11(1): 3252, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32591534

ABSTRACT

MiDAC is one of seven distinct, large multi-protein complexes that recruit class I histone deacetylases to the genome to regulate gene expression. Despite implications of involvement in cell cycle regulation and in several cancers, surprisingly little is known about the function or structure of MiDAC. Here we show that MiDAC is important for chromosome alignment during mitosis in cancer cell lines. Mice lacking the MiDAC proteins, DNTTIP1 or MIDEAS, die with identical phenotypes during late embryogenesis due to perturbations in gene expression that result in heart malformation and haematopoietic failure. This suggests that MiDAC has an essential and unique function that cannot be compensated by other HDAC complexes. Consistent with this, the cryoEM structure of MiDAC reveals a unique and distinctive mode of assembly. Four copies of HDAC1 are positioned at the periphery with outward-facing active sites suggesting that the complex may target multiple nucleosomes implying a processive deacetylase function.


Subject(s)
Embryonic Development , Histone Deacetylases/metabolism , Multiprotein Complexes/metabolism , Amino Acid Sequence , Animals , Cell Line , Chromatin/metabolism , Chromosomes, Mammalian/metabolism , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Gene Regulatory Networks , Heterozygote , Homozygote , Humans , Mice, Inbred C57BL , Mice, Knockout , Mitosis , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/ultrastructure , Nuclear Proteins/metabolism , Protein Domains , Protein Multimerization
17.
Elife ; 92020 06 05.
Article in English | MEDLINE | ID: mdl-32501215

ABSTRACT

Histone acetylation regulates chromatin structure and gene expression and is removed by histone deacetylases (HDACs). HDACs are commonly found in various protein complexes to confer distinct cellular functions, but how the multi-subunit complexes influence deacetylase activities and site-selectivities in chromatin is poorly understood. Previously we reported the results of studies on the HDAC1 containing CoREST complex and acetylated nucleosome substrates which revealed a notable preference for deacetylation of histone H3 acetyl-Lys9 vs. acetyl-Lys14 (Wu et al, 2018). Here we analyze the enzymatic properties of five class I HDAC complexes: CoREST, NuRD, Sin3B, MiDAC and SMRT with site-specific acetylated nucleosome substrates. Our results demonstrate that these HDAC complexes show a wide variety of deacetylase rates in a site-selective manner. A Gly13 in the histone H3 tail is responsible for a sharp reduction in deacetylase activity of the CoREST complex for H3K14ac. These studies provide a framework for connecting enzymatic and biological functions of specific HDAC complexes.


Subject(s)
Histone Deacetylases/metabolism , Histones/metabolism , Nucleosomes/metabolism , Acetylation , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Histone Deacetylases/genetics , Histones/genetics , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nucleosomes/genetics
18.
Chem Commun (Camb) ; 56(32): 4476-4479, 2020 Apr 21.
Article in English | MEDLINE | ID: mdl-32201871

ABSTRACT

We have identified a proteolysis targeting chimera (PROTAC) of class I HDACs 1, 2 and 3. The most active degrader consists of a benzamide HDAC inhibitor, an alkyl linker, and the von Hippel-Lindau E3 ligand. Our PROTAC increased histone acetylation levels and compromised colon cancer HCT116 cell viability, establishing a degradation strategy as an alternative to class I HDAC inhibition.


Subject(s)
Co-Repressor Proteins , Histone Deacetylases , Animals , Humans , Mice , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Co-Repressor Proteins/metabolism , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Histone Demethylases/antagonists & inhibitors , Proteolysis
19.
Cell Rep ; 30(8): 2699-2711.e8, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32101746

ABSTRACT

The transcriptional corepressor complex CoREST is one of seven histone deacetylase complexes that regulate the genome through controlling chromatin acetylation. The CoREST complex is unique in containing both histone demethylase and deacetylase enzymes, LSD1 and HDAC1, held together by the RCOR1 scaffold protein. To date, it has been assumed that the enzymes function independently within the complex. Now, we report the assembly of the ternary complex. Using both structural and functional studies, we show that the activity of the two enzymes is closely coupled and that the complex can exist in at least two distinct states with different kinetics. Electron microscopy of the complex reveals a bi-lobed structure with LSD1 and HDAC1 enzymes at opposite ends of the complex. The structure of CoREST in complex with a nucleosome reveals a mode of chromatin engagement that contrasts with previous models.


Subject(s)
Co-Repressor Proteins/metabolism , Histone Deacetylase 1/metabolism , Histone Demethylases/metabolism , Nerve Tissue Proteins/metabolism , Acetylation , Amino Acid Sequence , Animals , Cryoelectron Microscopy , Demethylation , HEK293 Cells , Humans , Kinetics , Models, Molecular , Nucleosomes/metabolism , Xenopus
20.
Urology ; 137: 178-182, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31866523

ABSTRACT

OBJECTIVE: To investigate the incidence and risk factors associated with artificial urinary sphincter (AUS) and male urethral sling placement (MUS), revision, and removal. METHODS: We identified CPT codes of patients undergoing AUS and sling placement, revision, and removal in the American College of Surgeons National Surgery Quality Improvement Program database. Bivariate analysis was used to compare preoperative parameters against adverse events of interest (Length of stay (LOS) >1, readmission, reoperation, other postoperative complications, and death). Variables that were significant or neared significance (P <.1) in the univariate analysis were entered into multivariable logistic regression models. Multivariable models were used to estimate the probability of adverse events. RESULTS: About 2792 patients underwent surgical treatment for stress urinary incontinence in the American College of Surgeons National Surgery Quality Improvement Program database from 2008 to 2016. Increased length of stay was the most common adverse event (12.7%), followed by other postoperative complications (4.9%), readmission (4%), reoperation (2.3%), and death (0.3%). We noted an association between perioperative adverse events and preoperative hypoalbuminemia. Patients with preoperative hypoalbuminemia compared with patients with normal preoperative serum albumin had an increase predicted probability of LOS >1 day (42% vs 10%), readmission (10% vs 4%), reoperation (6% vs 2%), other postoperative complications (18% vs 4%) after adjusting for other factors. CONCLUSION: Surgical treatment for stress urinary incontinence is well tolerated with acceptable levels of perioperative adverse events. Low serum albumin (<3.5 ng/dL) was associated with perioperative adverse events. These data may affect preoperative decision making and direct future quality improve efforts at the highest risk patients to help minimize perioperative morbidity and mortality.


Subject(s)
Postoperative Complications , Prosthesis Implantation , Reoperation , Serum Albumin/analysis , Suburethral Slings/statistics & numerical data , Urinary Incontinence, Stress , Urinary Sphincter, Artificial/statistics & numerical data , Aged , Humans , Length of Stay/statistics & numerical data , Male , Patient Readmission/statistics & numerical data , Postoperative Complications/diagnosis , Postoperative Complications/mortality , Postoperative Complications/surgery , Prosthesis Implantation/adverse effects , Prosthesis Implantation/instrumentation , Prosthesis Implantation/methods , Quality Improvement/organization & administration , Reoperation/methods , Reoperation/statistics & numerical data , Retrospective Studies , Risk Adjustment/methods , Risk Factors , United States/epidemiology , Urinary Incontinence, Stress/blood , Urinary Incontinence, Stress/epidemiology , Urinary Incontinence, Stress/surgery
SELECTION OF CITATIONS
SEARCH DETAIL
...