Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
Add more filters

Publication year range
1.
Cell ; 185(19): 3603-3616.e13, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36084631

ABSTRACT

The effects of mutations in continuously emerging variants of SARS-CoV-2 are a major concern for the performance of rapid antigen tests. To evaluate the impact of mutations on 17 antibodies used in 11 commercially available antigen tests with emergency use authorization, we measured antibody binding for all possible Nucleocapsid point mutations using a mammalian surface-display platform and deep mutational scanning. The results provide a complete map of the antibodies' epitopes and their susceptibility to mutational escape. Our data predict no vulnerabilities for detection of mutations found in variants of concern. We confirm this using the commercial tests and sequence-confirmed COVID-19 patient samples. The antibody escape mutational profiles generated here serve as a valuable resource for predicting the performance of rapid antigen tests against past, current, as well as any possible future variants of SARS-CoV-2, establishing the direct clinical and public health utility of our system.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Antibodies, Neutralizing , Antibodies, Viral , Epitopes/genetics , Humans , Mammals , Mutation , Nucleocapsid , SARS-CoV-2/genetics
2.
Nat Rev Mol Cell Biol ; 18(3): 159-174, 2017 03.
Article in English | MEDLINE | ID: mdl-28053348

ABSTRACT

The glucocorticoid receptor (GR) is a constitutively expressed transcriptional regulatory factor (TRF) that controls many distinct gene networks, each uniquely determined by particular cellular and physiological contexts. The precision of GR-mediated responses seems to depend on combinatorial, context-specific assembly of GR-nucleated transcription regulatory complexes at genomic response elements. In turn, evidence suggests that context-driven plasticity is conferred by the integration of multiple signals, each serving as an allosteric effector of GR conformation, a key determinant of regulatory complex composition and activity. This structural and mechanistic perspective on GR regulatory specificity is likely to extend to other eukaryotic TRFs.


Subject(s)
Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/metabolism , Acetylation , Animals , Chromatin Assembly and Disassembly , DNA/metabolism , Gene Expression Regulation , Humans , Phosphorylation , Protein Domains , Receptors, Glucocorticoid/genetics , Response Elements , Sumoylation , Transcription, Genetic
3.
Cell ; 159(1): 58-68, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25259920

ABSTRACT

Complex gene regulatory networks require transcription factors (TFs) to bind distinct DNA sequences. To understand how novel TF specificity evolves, we combined phylogenetic, biochemical, and biophysical approaches to interrogate how DNA recognition diversified in the steroid hormone receptor (SR) family. After duplication of the ancestral SR, three mutations in one copy radically weakened binding to the ancestral estrogen response element (ERE) and improved binding to a new set of DNA sequences (steroid response elements, SREs). They did so by establishing unfavorable interactions with ERE and abolishing unfavorable interactions with SRE; also required were numerous permissive substitutions, which nonspecifically improved cooperativity and affinity of DNA binding. Our findings indicate that negative determinants of binding play key roles in TFs' DNA selectivity and-with our prior work on the evolution of SR ligand specificity during the same interval-show how a specific new gene regulatory module evolved without interfering with the integrity of the ancestral module.


Subject(s)
Evolution, Molecular , Gene Regulatory Networks , Receptors, Steroid/chemistry , Receptors, Steroid/genetics , Response Elements , Animals , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Phylogeny , Receptors, Steroid/metabolism
4.
Nature ; 611(7937): 769-779, 2022 11.
Article in English | MEDLINE | ID: mdl-36385529

ABSTRACT

APOE4 is the strongest genetic risk factor for Alzheimer's disease1-3. However, the effects of APOE4 on the human brain are not fully understood, limiting opportunities to develop targeted therapeutics for individuals carrying APOE4 and other risk factors for Alzheimer's disease4-8. Here, to gain more comprehensive insights into the impact of APOE4 on the human brain, we performed single-cell transcriptomics profiling of post-mortem human brains from APOE4 carriers compared with non-carriers. This revealed that APOE4 is associated with widespread gene expression changes across all cell types of the human brain. Consistent with the biological function of APOE2-6, APOE4 significantly altered signalling pathways associated with cholesterol homeostasis and transport. Confirming these findings with histological and lipidomic analysis of the post-mortem human brain, induced pluripotent stem-cell-derived cells and targeted-replacement mice, we show that cholesterol is aberrantly deposited in oligodendrocytes-myelinating cells that are responsible for insulating and promoting the electrical activity of neurons. We show that altered cholesterol localization in the APOE4 brain coincides with reduced myelination. Pharmacologically facilitating cholesterol transport increases axonal myelination and improves learning and memory in APOE4 mice. We provide a single-cell atlas describing the transcriptional effects of APOE4 on the aging human brain and establish a functional link between APOE4, cholesterol, myelination and memory, offering therapeutic opportunities for Alzheimer's disease.


Subject(s)
Apolipoprotein E4 , Brain , Cholesterol , Nerve Fibers, Myelinated , Oligodendroglia , Animals , Humans , Mice , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Brain/metabolism , Brain/pathology , Cholesterol/metabolism , Oligodendroglia/metabolism , Oligodendroglia/pathology , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Autopsy , Induced Pluripotent Stem Cells , Neurons/metabolism , Neurons/pathology , Heterozygote , Biological Transport , Homeostasis , Single-Cell Analysis , Memory , Aging/genetics , Gene Expression Profiling , Myelin Sheath/metabolism , Myelin Sheath/pathology
5.
Immunity ; 49(1): 42-55.e6, 2018 07 17.
Article in English | MEDLINE | ID: mdl-30021146

ABSTRACT

The execution of shock following high dose E. coli lipopolysaccharide (LPS) or bacterial sepsis in mice required pro-apoptotic caspase-8 in addition to pro-pyroptotic caspase-11 and gasdermin D. Hematopoietic cells produced MyD88- and TRIF-dependent inflammatory cytokines sufficient to initiate shock without any contribution from caspase-8 or caspase-11. Both proteases had to be present to support tumor necrosis factor- and interferon-ß-dependent tissue injury first observed in the small intestine and later in spleen and thymus. Caspase-11 enhanced the activation of caspase-8 and extrinsic cell death machinery within the lower small intestine. Neither caspase-8 nor caspase-11 was individually sufficient for shock. Both caspases collaborated to amplify inflammatory signals associated with tissue damage. Therefore, combined pyroptotic and apoptotic signaling mediated endotoxemia independently of RIPK1 kinase activity and RIPK3 function. These observations bring to light the relevance of tissue compartmentalization to disease processes in vivo where cytokines act in parallel to execute diverse cell death pathways.


Subject(s)
Caspase 8/metabolism , Caspases/metabolism , Escherichia coli Infections/enzymology , Escherichia coli Infections/physiopathology , Shock, Septic/enzymology , Shock, Septic/physiopathology , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Caspase 8/genetics , Caspases/genetics , Caspases, Initiator , Cells, Cultured , Female , Inflammation/metabolism , Inflammation/pathology , Interferon Regulatory Factor-3/genetics , Interferon-beta/blood , Interferon-beta/metabolism , Intestine, Small/pathology , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides/toxicity , Male , Mice, Inbred C57BL , Mice, Knockout , Phosphate-Binding Proteins , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Spleen/pathology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism
6.
Nat Rev Mol Cell Biol ; 15(11): 749-60, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25269475

ABSTRACT

Proteins that bind both DNA and RNA typify the ability of a single gene product to perform multiple functions. Such DNA- and RNA-binding proteins (DRBPs) have unique functional characteristics that stem from their specific structural features; these developed early in evolution and are widely conserved. Proteins that bind RNA have typically been considered as functionally distinct from proteins that bind DNA and studied independently. This practice is becoming outdated, in partly owing to the discovery of long non-coding RNAs (lncRNAs) that target DNA-binding proteins. Consequently, DRBPs were found to regulate many cellular processes, including transcription, translation, gene silencing, microRNA biogenesis and telomere maintenance.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Evolution, Molecular , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , Animals , Binding, Competitive , DNA/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Databases, Genetic , Gene Silencing , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Models, Molecular , Protein Binding , Protein Biosynthesis , RNA, Long Noncoding/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Telomere Homeostasis , Transcription, Genetic
7.
J Biol Chem ; : 107855, 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39369989

ABSTRACT

Thioesterase superfamily member 2 (Them2), a long-chain fatty acyl-CoA thioesterase that is highly expressed in oxidative tissues, interacts with phosphatidylcholine transfer protein (PC-TP) to regulate hepatic lipid and glucose metabolism and to suppress insulin signaling. High-fat diet (HFD)-fed mice lacking Them2 globally or specifically in skeletal muscle, but not liver, exhibit reduced hepatic steatosis and insulin resistance. Here, we report that the capacity of Them2 in skeletal muscle to promote hepatic steatosis and insulin resistance depends on both its catalytic activity and interaction with PC-TP. Two residues of Them2 catalytic site were mutated (N50A/D65A) to produce the inactive enzyme while maintaining its homotetrameric structure and interaction with PC-TP. Restoration of skeletal muscle expression in Them2-/- mice using recombinant adeno-associated virus revealed that wild-type (WT), but not N50A/D65A Them2, promoted HFD-induced weight gain and hepatic steatosis. This was accompanied by greater impairment of insulin sensitivity in WT compared with N50A/D65A Them2. Pharmacological inhibition or genetic ablation of PC-TP attenuated these effects. In reductionist experiments, conditioned medium collected from WT primary cultured myotubes promoted excess lipid accumulation in oleic acid-treated primary cultured hepatocytes relative to Them2-/- myotubes, which was attributable to secreted extracellular vesicles (EV). Reconstitution of Them2 expression in Them2-/- myotubes affirmed the requirements for catalytic activity and PC-TP interactions for EV to promote lipid accumulation in hepatocytes. These studies provide valuable mechanistic insights whereby Them2 in skeletal muscle promotes hepatic steatosis and establish both Them2 and PC-TP as represent attractive targets for managing metabolic dysfunction-associated steatotic liver disease.

8.
Nucleic Acids Res ; 51(15): 7972-7987, 2023 08 25.
Article in English | MEDLINE | ID: mdl-37395399

ABSTRACT

DNA-dependent protein kinase (DNA-PK) plays a critical role in non-homologous end joining (NHEJ), the predominant pathway that repairs DNA double-strand breaks (DSB) in response to ionizing radiation (IR) to govern genome integrity. The interaction of the catalytic subunit of DNA-PK (DNA-PKcs) with the Ku70/Ku80 heterodimer on DSBs leads to DNA-PK activation; however, it is not known if upstream signaling events govern this activation. Here, we reveal a regulatory step governing DNA-PK activation by SIRT2 deacetylation, which facilitates DNA-PKcs localization to DSBs and interaction with Ku, thereby promoting DSB repair by NHEJ. SIRT2 deacetylase activity governs cellular resistance to DSB-inducing agents and promotes NHEJ. SIRT2 furthermore interacts with and deacetylates DNA-PKcs in response to IR. SIRT2 deacetylase activity facilitates DNA-PKcs interaction with Ku and localization to DSBs and promotes DNA-PK activation and phosphorylation of downstream NHEJ substrates. Moreover, targeting SIRT2 with AGK2, a SIRT2-specific inhibitor, augments the efficacy of IR in cancer cells and tumors. Our findings define a regulatory step for DNA-PK activation by SIRT2-mediated deacetylation, elucidating a critical upstream signaling event initiating the repair of DSBs by NHEJ. Furthermore, our data suggest that SIRT2 inhibition may be a promising rationale-driven therapeutic strategy for increasing the effectiveness of radiation therapy.


Subject(s)
DNA Breaks, Double-Stranded , Protein Kinases , DNA/genetics , DNA/metabolism , DNA End-Joining Repair , DNA Repair , DNA-Activated Protein Kinase/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Ku Autoantigen/metabolism , Nuclear Proteins/metabolism , Protein Kinases/genetics , Sirtuin 2/genetics , Sirtuin 2/metabolism , Humans
9.
J Biol Chem ; 299(8): 104921, 2023 08.
Article in English | MEDLINE | ID: mdl-37328104

ABSTRACT

Steroidogenic factor-1 (SF-1) is a phospholipid-sensing nuclear receptor expressed in the adrenal glands, gonads, and hypothalamus which controls steroidogenesis and metabolism. There is significant therapeutic interest in SF-1 because of its oncogenic properties in adrenocortical cancer. Synthetic modulators are attractive for targeting SF-1 for clinical and laboratory purposes due to the poor pharmaceutical properties of its native phospholipid ligands. While small molecule agonists targeting SF-1 have been synthesized, no crystal structures have been reported of SF-1 in complexes with synthetic compounds. This has prevented the establishment of structure-activity relationships that would enable better characterization of ligand-mediated activation and improvement in current chemical scaffolds. Here, we compare the effects of small molecules in SF-1 and its close homolog, liver receptor homolog-1 (LRH-1), and identify several molecules that specifically activate LRH-1. We also report the first crystal structure of SF-1 in complex with a synthetic agonist that displays low nanomolar affinity and potency for SF-1. We use this structure to explore the mechanistic basis for small molecule agonism of SF-1, especially compared to LRH-1, and uncover unique signaling pathways that drive LRH-1 specificity. Molecular dynamics simulations reveal differences in protein dynamics at the pocket mouth as well as ligand-mediated allosteric communication from this region to the coactivator binding interface. Our studies, therefore, shed important insight into the allostery driving SF-1 activity and show potential for modulation of LRH-1 over SF-1.


Subject(s)
Models, Molecular , Molecular Dynamics Simulation , Receptors, Cytoplasmic and Nuclear , Small Molecule Libraries , Steroidogenic Factor 1 , Ligands , Phospholipids/chemistry , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/chemistry , Small Molecule Libraries/chemistry , Steroidogenic Factor 1/agonists , Steroidogenic Factor 1/chemistry , Humans , Crystallography, X-Ray
10.
Biochem Soc Trans ; 52(4): 1565-1577, 2024 08 28.
Article in English | MEDLINE | ID: mdl-39140379

ABSTRACT

Lipid synthesis and transport are essential for energy, production of cell membrane, and cell signaling. Acyl-CoA thioesterases (ACOTs) function to regulate intracellular levels of fatty acyl-CoAs through hydrolysis. Two members of this family, ACOT11 and ACOT12, contain steroidogenic acute regulatory related lipid transfer domains, which typically function as lipid transport or regulatory domains. This work reviews ACOT11 and ACOT12 structures and functions, and the potential role of the START domains in lipid transfer activity and the allosteric regulation of catalytic activity.


Subject(s)
Thiolester Hydrolases , Thiolester Hydrolases/metabolism , Thiolester Hydrolases/chemistry , Humans , Lipid Metabolism , Animals , Models, Molecular , Allosteric Regulation
11.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Article in English | MEDLINE | ID: mdl-34285072

ABSTRACT

The glucocorticoid receptor (GR) is a ligand-regulated transcription factor (TF) that controls the tissue- and gene-specific transactivation and transrepression of thousands of target genes. Distinct GR DNA-binding sequences with activating or repressive activities have been identified, but how they modulate transcription in opposite ways is not known. We show that GR forms phase-separated condensates that specifically concentrate known coregulators via their intrinsically disordered regions (IDRs) in vitro. A combination of dynamic, multivalent (between IDRs) and specific, stable interactions (between LxxLL motifs and the GR ligand-binding domain) control the degree of recruitment. Importantly, GR DNA binding directs the selective partitioning of coregulators within GR condensates such that activating DNAs cause enhanced recruitment of coactivators. Our work shows that condensation controls GR function by modulating coregulator recruitment and provides a mechanism for the up- and down-regulation of GR target genes controlled by distinct DNA recognition elements.


Subject(s)
Gene Expression Regulation , Promoter Regions, Genetic , Protein Multimerization , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/metabolism , Regulatory Sequences, Nucleic Acid , Transcriptional Activation , Glucocorticoids/metabolism , Humans
12.
J Clin Microbiol ; 61(10): e0013823, 2023 10 24.
Article in English | MEDLINE | ID: mdl-37728336

ABSTRACT

Rapid antigen tests (RATs) have become an invaluable tool for combating the COVID-19 pandemic. However, concerns have been raised regarding the ability of existing RATs to effectively detect emerging SARS-CoV-2 variants. We compared the performance of 10 commercially available, emergency use authorized RATs against the Delta and Omicron SARS-CoV-2 variants using both individual patient and serially diluted pooled clinical samples. The RATs exhibited lower sensitivity for Omicron samples when using PCR cycle threshold (CT) value (a rough proxy for RNA concentration) as the comparator. Interestingly, however, they exhibited similar sensitivity for Omicron and Delta samples when using quantitative antigen concentration as the comparator. We further found that the Omicron samples had lower ratios of antigen to RNA, which offers a potential explanation for the apparent lower sensitivity of RATs for that variant when using C T value as a reference. Our findings underscore the complexity in assessing RAT performance against emerging variants and highlight the need for ongoing evaluation in the face of changing population immunity and virus evolution.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , COVID-19/diagnosis , Pandemics , RNA
13.
PLoS Pathog ; 17(9): e1009941, 2021 09.
Article in English | MEDLINE | ID: mdl-34559866

ABSTRACT

The metabolic signaling pathways that drive pathologic tissue inflammation and damage in humans with pulmonary tuberculosis (TB) are not well understood. Using combined methods in plasma high-resolution metabolomics, lipidomics and cytokine profiling from a multicohort study of humans with pulmonary TB disease, we discovered that IL-1ß-mediated inflammatory signaling was closely associated with TCA cycle remodeling, characterized by accumulation of the proinflammatory metabolite succinate and decreased concentrations of the anti-inflammatory metabolite itaconate. This inflammatory metabolic response was particularly active in persons with multidrug-resistant (MDR)-TB that received at least 2 months of ineffective treatment and was only reversed after 1 year of appropriate anti-TB chemotherapy. Both succinate and IL-1ß were significantly associated with proinflammatory lipid signaling, including increases in the products of phospholipase A2, increased arachidonic acid formation, and metabolism of arachidonic acid to proinflammatory eicosanoids. Together, these results indicate that decreased itaconate and accumulation of succinate and other TCA cycle intermediates is associated with IL-1ß-mediated proinflammatory eicosanoid signaling in pulmonary TB disease. These findings support host metabolic remodeling as a key driver of pathologic inflammation in human TB disease.


Subject(s)
Citric Acid Cycle/physiology , Inflammation/metabolism , Signal Transduction/physiology , Tuberculosis, Pulmonary/metabolism , Humans
14.
Hepatology ; 75(1): 154-169, 2022 01.
Article in English | MEDLINE | ID: mdl-34433228

ABSTRACT

BACKGROUND AND AIMS: Thioesterase superfamily member 2 (Them2) is highly expressed in liver and oxidative tissues, where it hydrolyzes long-chain fatty acyl-CoA esters to free fatty acids and CoA. Although mice globally lacking Them2 (Them2-/- ) are protected against diet-induced obesity, hepatic steatosis (HS), and insulin resistance (IR), liver-specific Them2-/- mice remain susceptible. The aim of this study was to test whether Them2 activity in extrahepatic oxidative tissues is a primary determinant of HS and IR. APPROACH AND RESULTS: Upon observing IR and up-regulation of Them2 in skeletal, but not cardiac, muscle of high-fat-diet (HFD)-fed wild-type compared to Them2-/- mice, we created mice with Them2 specifically deleted in skeletal (S-Them2-/- ) and cardiac muscle (C-Them2-/- ), as well as in adipose tissue (A-Them2-/- ). When fed an HFD, S-Them2-/- , but not C-Them2-/- or A-Them2-/- , mice exhibited reduced weight gain and improved glucose homeostasis and insulin sensitivity. Reconstitution of Them2 expression in skeletal muscle of global Them2-/- mice, using adeno-associated virus, was sufficient to restore excess weight gain. Increased rates of fatty acid oxidation in skeletal muscle of S-Them2-/- mice contributed to protection from HFD-induced HS by increasing VLDL triglyceride secretion rates in response to greater demand. Increases in insulin sensitivity were further attributable to alterations in production of skeletal muscle metabolites, including short-chain fatty acids, branched-chain amino acids, and pentose phosphate pathway intermediates, as well as in expression of myokines that modulate insulin responsiveness. CONCLUSIONS: These results reveal a key role for skeletal muscle Them2 in the pathogenesis of HS and IR and implicate it as a target in the management of NAFLD.


Subject(s)
Insulin Resistance/genetics , Lipid Metabolism/genetics , Muscle, Skeletal/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Thiolester Hydrolases/metabolism , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Humans , Male , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Oxidation-Reduction , Thiolester Hydrolases/genetics , Up-Regulation
15.
Nat Chem Biol ; 17(3): 307-316, 2021 03.
Article in English | MEDLINE | ID: mdl-33510451

ABSTRACT

Glucocorticoids display remarkable anti-inflammatory activity, but their use is limited by on-target adverse effects including insulin resistance and skeletal muscle atrophy. We used a chemical systems biology approach, ligand class analysis, to examine ligands designed to modulate glucocorticoid receptor activity through distinct structural mechanisms. These ligands displayed diverse activity profiles, providing the variance required to identify target genes and coregulator interactions that were highly predictive of their effects on myocyte glucose disposal and protein balance. Their anti-inflammatory effects were linked to glucose disposal but not muscle atrophy. This approach also predicted selective modulation in vivo, identifying compounds that were muscle-sparing or anabolic for protein balance and mitochondrial potential. Ligand class analysis defined the mechanistic links between the ligand-receptor interface and ligand-driven physiological outcomes, a general approach that can be applied to any ligand-regulated allosteric signaling system.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Glucose Transporter Type 4/genetics , Muscular Atrophy/drug therapy , Receptors, Glucocorticoid/chemistry , Signal Transduction/drug effects , A549 Cells , Allosteric Regulation , Animals , Anti-Inflammatory Agents/chemical synthesis , Cell Line, Transformed , Gene Expression Regulation , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Humans , Lipopolysaccharides/administration & dosage , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscular Atrophy/chemically induced , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Myoblasts/drug effects , Myoblasts/metabolism , Rats , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Structure-Activity Relationship
16.
Nucleic Acids Res ; 49(15): 8923-8933, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34289059

ABSTRACT

The most common form of DNA methylation involves the addition of a methyl group to a cytosine base in the context of a cytosine-phosphate-guanine (CpG) dinucleotide. Genomes from more primitive organisms are more abundant in CpG sites that, through the process of methylation, deamination and subsequent mutation to thymine-phosphate-guanine (TpG) sites, can produce new transcription factor binding sites. Here, we examined the evolutionary history of the over 36 000 glucocorticoid receptor (GR) consensus binding motifs in the human genome and identified a subset of them in regulatory regions that arose via a deamination and subsequent mutation event. GR can bind to both unmodified and methylated pre-GR binding sequences (GBSs) that contain a CpG site. Our structural analyses show that CpG methylation in a pre-GBS generates a favorable interaction with Arg447 mimicking that made with a TpG in a GBS. This methyl-specific recognition arose 420 million years ago and was conserved during the evolution of GR and likely helps fix the methylation on the relevant cytosines. Our study provides the first genetic, biochemical and structural evidence of high-affinity binding for the likely evolutionary precursor of extant TpG-containing GBS.


Subject(s)
DNA Methylation/genetics , Evolution, Molecular , Genome, Human/genetics , Receptors, Glucocorticoid/genetics , Binding Sites/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/ultrastructure , Dinucleoside Phosphates/genetics , Humans , Nucleic Acid Conformation , Receptors, Glucocorticoid/ultrastructure , Regulatory Sequences, Nucleic Acid/genetics , Thymine/chemistry
17.
Proc Natl Acad Sci U S A ; 117(39): 24285-24293, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32917814

ABSTRACT

Duchenne muscular dystrophy is a genetic disorder that shows chronic and progressive damage to skeletal and cardiac muscle leading to premature death. Antiinflammatory corticosteroids targeting the glucocorticoid receptor (GR) are the current standard of care but drive adverse side effects such as deleterious bone loss. Through subtle modification to a steroidal backbone, a recently developed drug, vamorolone, appears to preserve beneficial efficacy but with significantly reduced side effects. We use combined structural, biophysical, and biochemical approaches to show that loss of a receptor-ligand hydrogen bond drives these remarkable therapeutic effects. Moreover, vamorolone uniformly weakens coactivator associations but not corepressor associations, implicating partial agonism as the main driver of its dissociative properties. Additionally, we identify a critical and evolutionarily conserved intramolecular network connecting the ligand to the coregulator binding surface. Interruption of this allosteric network by vamorolone selectively reduces GR-driven transactivation while leaving transrepression intact. Our results establish a mechanistic understanding of how vamorolone reduces side effects, guiding the future design of partial agonists as selective GR modulators with an improved therapeutic index.


Subject(s)
Muscular Dystrophy, Duchenne/drug therapy , Pregnadienediols/administration & dosage , Pregnadienediols/chemistry , Humans , Hydrogen Bonding , Ligands , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Protein Binding , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism
18.
Proc Natl Acad Sci U S A ; 117(35): 21673-21680, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32817554

ABSTRACT

Behavioral evolution relies on genetic changes, yet few behaviors can be traced to specific genetic sequences in vertebrates. Here we provide experimental evidence showing that differentiation of a single gene has contributed to the evolution of divergent behavioral phenotypes in the white-throated sparrow, a common backyard songbird. In this species, a series of chromosomal inversions has formed a supergene that segregates with an aggressive phenotype. The supergene has captured ESR1, the gene that encodes estrogen receptor α (ERα); as a result, this gene is accumulating changes that now distinguish the supergene allele from the standard allele. Our results show that in birds of the more aggressive phenotype, ERα knockdown caused a phenotypic change to that of the less aggressive phenotype. We next showed that in a free-living population, aggression is predicted by allelic imbalance favoring the supergene allele. Finally, we identified cis-regulatory features, both genetic and epigenetic, that explain the allelic imbalance. This work provides a rare illustration of how genotypic divergence has led to behavioral phenotypic divergence in a vertebrate.


Subject(s)
Aggression/physiology , Estrogen Receptor alpha/genetics , Sparrows/genetics , Animals , Behavior, Animal , Chromosome Inversion/genetics , Estrogens/genetics , Estrogens/metabolism , Female , Genotype , Male , Phenotype , Receptors, Estrogen/genetics , Social Behavior
19.
Proc Natl Acad Sci U S A ; 117(36): 22080-22089, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32820071

ABSTRACT

Nonshivering thermogenesis occurs in brown adipose tissue to generate heat in response to cold ambient temperatures. Thioesterase superfamily member 1 (Them1) is transcriptionally up-regulated in brown adipose tissue upon exposure to the cold and suppresses thermogenesis in order to conserve energy reserves. It hydrolyzes long-chain fatty acyl-CoAs that are derived from lipid droplets, preventing their use as fuel for thermogenesis. In addition to its enzymatic domains, Them1 contains a C-terminal StAR-related lipid transfer (START) domain with unknown ligand or function. By complementary biophysical approaches, we show that the START domain binds to long-chain fatty acids, products of Them1's enzymatic reaction, as well as lysophosphatidylcholine (LPC), lipids shown to activate thermogenesis in brown adipocytes. Certain fatty acids stabilize the START domain and allosterically enhance Them1 catalysis of acyl-CoA, whereas 18:1 LPC destabilizes and inhibits activity, which we verify in cell culture. Additionally, we demonstrate that the START domain functions to localize Them1 near lipid droplets. These findings define the role of the START domain as a lipid sensor that allosterically regulates Them1 activity and spatially localizes it in proximity to the lipid droplet.


Subject(s)
Fatty Acids/metabolism , Lysophosphatidylcholines/metabolism , Palmitoyl-CoA Hydrolase/chemistry , Palmitoyl-CoA Hydrolase/metabolism , Acyl Coenzyme A/metabolism , Adipose Tissue, Brown/enzymology , Adipose Tissue, Brown/metabolism , Allosteric Regulation , Fatty Acids/chemistry , Humans , Kinetics , Lipid Droplets/enzymology , Lipid Droplets/metabolism , Lysophosphatidylcholines/chemistry , Palmitoyl-CoA Hydrolase/genetics , Protein Domains
20.
J Biol Chem ; 296: 100024, 2021.
Article in English | MEDLINE | ID: mdl-33410399

ABSTRACT

The human genome contains vast genetic diversity as naturally occurring coding variants, yet the impact of these variants on protein function and physiology is poorly understood. RGS14 is a multifunctional signaling protein that suppresses synaptic plasticity in dendritic spines of hippocampal neurons. RGS14 also is a nucleocytoplasmic shuttling protein, suggesting that balanced nuclear import/export and dendritic spine localization are essential for RGS14 functions. We identified genetic variants L505R (LR) and R507Q (RQ) located within the nuclear export sequence (NES) of human RGS14. Here we report that RGS14 encoding LR or RQ profoundly impacts protein functions in hippocampal neurons. RGS14 membrane localization is regulated by binding Gαi-GDP, whereas RGS14 nuclear export is regulated by Exportin 1 (XPO1). Remarkably, LR and RQ variants disrupt RGS14 binding to Gαi1-GDP and XPO1, nucleocytoplasmic equilibrium, and capacity to inhibit long-term potentiation (LTP). Variant LR accumulates irreversibly in the nucleus, preventing RGS14 binding to Gαi1, localization to dendritic spines, and inhibitory actions on LTP induction, while variant RQ exhibits a mixed phenotype. When introduced into mice by CRISPR/Cas9, RGS14-LR protein expression was detected predominantly in the nuclei of neurons within hippocampus, central amygdala, piriform cortex, and striatum, brain regions associated with learning and synaptic plasticity. Whereas mice completely lacking RGS14 exhibit enhanced spatial learning, mice carrying variant LR exhibit normal spatial learning, suggesting that RGS14 may have distinct functions in the nucleus independent from those in dendrites and spines. These findings show that naturally occurring genetic variants can profoundly alter normal protein function, impacting physiology in unexpected ways.


Subject(s)
Cell Nucleus/metabolism , Hippocampus/metabolism , Long-Term Potentiation , Mutation , Neurons/metabolism , RGS Proteins/genetics , Animals , Hippocampus/cytology , Hippocampus/physiology , Humans , Karyopherins/metabolism , Mice , Neuronal Plasticity , Protein Transport , RGS Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Spatial Learning , Exportin 1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL