Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Am J Med Genet A ; 188(2): 556-568, 2022 02.
Article in English | MEDLINE | ID: mdl-34726335

ABSTRACT

Predicting genotype-to-phenotype correlations from genomic variants has been challenging, particularly for genes that have a complex balance of dominant and recessive inheritance for phenotypes. Variants in NMDA receptor components GRIN1, GRIN2A, and GRIN2B cause a myriad of dominant disease phenotypes, with the most common being epilepsy and autism spectrum disorder. Starting from the analysis of a variant of uncertain significance (VUS, GRIN2A G760S), we realized the need for tools to map dominant variants for the components of the NMDA receptor. Some variants within GRIN1, GRIN2A, and GRIN2B exert dominant epilepsy and developmental delay, yet other amino acid variants are conserved and predicted to alter protein function but do not have dominant phenotypes. Common variant annotation tools are not powered to determine pathogenic dominant outcomes. To address this gap, we integrated sequence and structural analyses for GRIN1, GRIN2A, and GRIN2B. Using this approach, we determined that paralog homology mapping and topology can segregate dominant variants, with an elevation of intermolecular contacts between the subunits. Furthermore, demonstrating the general utility of our methodology, we show that 25 VUS within ClinVar also reach a dominant variant annotation, including the GRIN2A G760S variant. Our work suggests paralog homology and protein topology as a powerful strategy within the receptor complex to resolve dominant genetic variants relative to variants that would fit a recessive inheritance, requiring two damaging variants. These strategies should be tested in additional dominant genetic disorders to determine the broader utility.


Subject(s)
Autism Spectrum Disorder , Epilepsy , Epilepsy/genetics , Humans , N-Methylaspartate/genetics , Phenotype , Receptors, N-Methyl-D-Aspartate/genetics
2.
Genes (Basel) ; 12(4)2021 03 25.
Article in English | MEDLINE | ID: mdl-33806076

ABSTRACT

Ornithine decarboxylase 1 (ODC1 gene) has been linked through gain-of-function variants to a rare disease featuring developmental delay, alopecia, macrocephaly, and structural brain anomalies. ODC1 has been linked to additional diseases like cancer, with growing evidence for neurological contributions to schizophrenia, mood disorders, anxiety, epilepsy, learning, and suicidal behavior. The evidence of ODC1 connection to neural disorders highlights the need for a systematic analysis of ODC1 genotype-to-phenotype associations. An analysis of variants from ClinVar, Geno2MP, TOPMed, gnomAD, and COSMIC revealed an intellectual disability and seizure connected loss-of-function variant, ODC G84R (rs138359527, NC_000002.12:g.10444500C > T). The missense variant is found in ~1% of South Asian individuals and results in 2.5-fold decrease in enzyme function. Expression quantitative trait loci (eQTLs) reveal multiple functionally annotated, non-coding variants regulating ODC1 that associate with psychiatric/neurological phenotypes. Further dissection of RNA-Seq during fetal brain development and within cerebral organoids showed an association of ODC1 expression with cell proliferation of neural progenitor cells, suggesting gain-of-function variants with neural over-proliferation and loss-of-function variants with neural depletion. The linkage from the expression data of ODC1 in early neural progenitor proliferation to phenotypes of neurodevelopmental delay and to the connection of polyamine metabolites in brain function establish ODC1 as a bona fide neurodevelopmental disorder gene.


Subject(s)
Brain/pathology , Dicarboxylic Acid Transporters/genetics , Mitochondrial Membrane Transport Proteins/genetics , Neural Stem Cells/pathology , Neurodevelopmental Disorders/pathology , Phenotype , Polymorphism, Single Nucleotide , Brain/metabolism , Cell Proliferation , Humans , Neural Stem Cells/metabolism , Neurodevelopmental Disorders/genetics
3.
Front Mol Neurosci ; 13: 573409, 2020.
Article in English | MEDLINE | ID: mdl-33071758

ABSTRACT

The TSC1 and TSC2 genes are connected to multiple syndromes from Tuberous Sclerosis Complex (TSC) to autism spectrum disorder (ASD), with uncertainty if genetic variants cause all or subsets of phenotypes based on the location and type of change. For TSC1, few have addressed if non-TSC associated genetic variants have direct contributions to changes in neurological genotype-to-phenotype impacts, including elevated rates of ASD and seizures. Dominant variants cause TSC, yet TSC1 has many heritable variants not dominant for TSC that are poorly understood in neurological function, with some associated with ASD. Herein, we examined how missense variants in TSC1, R336W, T360N, T393I, S403L, and H732Y, impacted the development of cortical inhibitory interneurons, cell-types whose molecular, cellular, and physiological properties are altered after the loss of mouse TSC1. We found these variants complemented a known phenotype caused by loss of TSC1, increased cell size. However, distinct variants, particularly S403L showed deficits in complementing an increase in parvalbumin levels and exhibited smaller amplitude after hyperpolarizations. Overall, these data show that subtle phenotypes can be induced by some TSC1 missense variants and provide an in vivo system to assess TSC1 variants' neurological impact better.

4.
Article in English | MEDLINE | ID: mdl-33050454

ABSTRACT

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by disruptions in social communication and behavioral flexibility. Both genetic and environmental factors contribute to ASD risk. Epidemiologic studies indicate that roadway vehicle exhaust and in utero exposure to diesel particulate matter (DPM) are associated with ASD. Using the Comparative Toxicogenomics Database (CTD), we identified genes connected to DPM exposure and ASD, extracted the known enhancers/promoters of the identified genes, and integrated this with Assay for Transposase Accessible Chromatin (ATAC-seq) data from DPM-exposed human neural progenitor cells. Enhancer/promoter elements with significantly different chromosome accessibility revealed enriched DNA sequence motifs with transcription factor binding sites for EGR1. Variant extraction for linkage disequilibrium blocks of these regions followed by analysis through Genome Wide Association Studies (GWAS) revealed multiple neurological trait associations including exploratory eye movement and brain volume measurement. This approach highlights the effects of pollution on the regulatory regions of genes implicated in ASD by genetic studies, indicating convergence of genetic and environmental factors on molecular networks that contribute to ASD. Integration of publicly available data from the CTD, cell culture exposure studies, and phenotypic genetics synergize extensive evidence of chemical exposures on gene regulation for altered brain development.


Subject(s)
Autism Spectrum Disorder , Environmental Pollutants , Epigenesis, Genetic , Particulate Matter , Toxicogenetics , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/genetics , Environmental Pollutants/toxicity , Epigenesis, Genetic/drug effects , Female , Genome-Wide Association Study , Humans , Maternal Exposure , Particulate Matter/toxicity
5.
Proc Natl Acad Sci U S A ; 117(11): 6189-6195, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32123116

ABSTRACT

Neurofibromatosis 1 (NF1) is caused by mutations in the NF1 gene, which encodes the protein, neurofibromin, an inhibitor of Ras activity. Cortical GABAergic interneurons (CINs) are implicated in NF1 pathology, but the cellular and molecular changes to CINs are unknown. We deleted mouse Nf1 from the medial ganglionic eminence, which gives rise to both oligodendrocytes and CINs that express somatostatin and parvalbumin. Nf1 loss led to a persistence of immature oligodendrocytes that prevented later-generated oligodendrocytes from occupying the cortex. Moreover, molecular and cellular properties of parvalbumin (PV)-positive CINs were altered by the loss of Nf1, without changes in somatostatin (SST)-positive CINs. We discovered that loss of Nf1 results in a dose-dependent decrease in Lhx6 expression, the transcription factor necessary to establish SST+ and PV+ CINs, which was rescued by the MEK inhibitor SL327, revealing a mechanism whereby a neurofibromin/Ras/MEK pathway regulates a critical CIN developmental milestone.


Subject(s)
Cerebral Cortex/pathology , GABAergic Neurons/pathology , Interneurons/pathology , LIM-Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurofibromatosis 1/pathology , Neurofibromin 1/genetics , Transcription Factors/metabolism , Aminoacetonitrile/administration & dosage , Aminoacetonitrile/analogs & derivatives , Animals , Cells, Cultured , Cerebral Cortex/cytology , Disease Models, Animal , Embryo, Mammalian , Female , GABAergic Neurons/metabolism , Humans , Interneurons/metabolism , MAP Kinase Signaling System/drug effects , Median Eminence/cytology , Mice , Mice, Knockout , Neurofibromatosis 1/genetics , Neurofibromin 1/metabolism , Neuroglia/cytology , Parvalbumins/metabolism , Primary Cell Culture , Somatostatin/metabolism , ras GTPase-Activating Proteins/metabolism
6.
Epigenetics Chromatin ; 13(1): 18, 2020 03 16.
Article in English | MEDLINE | ID: mdl-32178718

ABSTRACT

BACKGROUND: MeCP2 and MBD2 are members of a family of proteins that possess a domain that selectively binds 5-methylcytosine in a CpG context. Members of the family interact with other proteins to modulate DNA packing. Stretching of DNA-protein complexes in nanofluidic channels with a cross-section of a few persistence lengths allows us to probe the degree of compaction by proteins. RESULTS: We demonstrate DNA compaction by MeCP2 while MBD2 does not affect DNA configuration. By using atomic force microscopy (AFM), we determined that the mechanism for compaction by MeCP2 is the formation of bridges between distant DNA stretches and the formation of loops. CONCLUSIONS: Despite sharing a similar specific DNA-binding domain, the impact of full-length 5-methylcytosine-binding proteins can vary drastically between strong compaction of DNA and no discernable large-scale impact of protein binding. We demonstrate that ATTO 565-labeled MBD2 is a good candidate as a staining agent for epigenetic mapping.


Subject(s)
5-Methylcytosine/metabolism , DNA Methylation , DNA-Binding Proteins/metabolism , DNA/chemistry , Methyl-CpG-Binding Protein 2/metabolism , Microfluidics/methods , 5-Methylcytosine/chemistry , Binding Sites , DNA/metabolism , DNA-Binding Proteins/chemistry , Epigenomics/methods , Humans , Methyl-CpG-Binding Protein 2/chemistry , Microfluidics/instrumentation , Microscopy, Atomic Force/methods , Protein Binding
7.
Dev Neurosci ; 42(5-6): 195-207, 2020.
Article in English | MEDLINE | ID: mdl-33657557

ABSTRACT

Autism spectrum disorder (ASD) manifests early in childhood. While genetic variants increase risk for ASD, a growing body of literature has established that in utero chemical exposures also contribute to ASD risk. These chemicals include air-based pollutants like diesel particulate matter (DPM). A combination of single-cell and direct transcriptomics of DPM-exposed human-induced pluripotent stem cell-derived cerebral organoids revealed toxicogenomic effects of DPM exposure during fetal brain development. Direct transcriptomics, sequencing RNA bases via Nanopore, revealed that cerebral organoids contain extensive RNA modifications, with DPM-altering cytosine methylation in oxidative mitochondrial transcripts expressed in outer radial glia cells. Single-cell transcriptomics further confirmed an oxidative phosphorylation change in cell groups such as outer radial glia upon DPM exposure. This approach highlights how DPM exposure perturbs normal mitochondrial function and cellular respiration during early brain development, which may contribute to developmental disorders like ASD by altering neurodevelopment.


Subject(s)
Epigenesis, Genetic/drug effects , Neurogenesis/drug effects , Particulate Matter/toxicity , Pluripotent Stem Cells/drug effects , Vehicle Emissions/toxicity , Autism Spectrum Disorder/etiology , Brain/drug effects , Female , Humans , Maternal Exposure/adverse effects , Organoids , Sequence Analysis, RNA
8.
Front Genet ; 10: 970, 2019.
Article in English | MEDLINE | ID: mdl-31681417

ABSTRACT

Most of the genetic risk for autism spectrum disorder (ASD) is inherited as common genetic variants, although some rare mutations have been identified in individuals with ASD. Common genetic variants are most parsimoniously identified by genome wide association studies. Genome wide association studies have identified several genetic loci with genome wide association with ASD. However, genome wide association studies only identify regions of the genome associated with phenotypic traits. Identification of the functional elements requires additional experimental evidence. Here, we demonstrate that a genome wide association study locus for ASD on chromosome 20p12.1, rs4141463, implicates a noncoding RNA as a functional element. Although rs4141463 lies within an intron of the protein-coding MACROD2 (MACRO domain containing 2) gene, expression of MACROD2 is neither altered in postmortem temporal cortex of individuals with ASD nor correlated with rs4141463 genotype. Our bioinformatics approaches revealed a noncoding RNA transcript near the autism susceptibility signal, RPS10P2-AS1 (ribosomal protein S10 pseudogene 2 anti-sense 1). In a panel of 15 human tissues, RPS10P2-AS1 was expressed at higher levels than the protein-coding MACROD2 in both fetal temporal cortex and adult peripheral blood. In postmortem temporal cortex, expression of RPS10P2-AS1 was increased 7-fold in individuals with ASD (P = 0.02) and increased 8-fold in individuals with the ASD-associated rs4141463 genotype (P = 0.01). Further, RPS10P2-AS1 expression was increased in human neural progenitor cells exposed to model air pollutants, indicating that both genetic and environmental factors that contribute to ASD increased RPS10P2-AS1 expression. Overexpression of RPS10P2-AS1 in human neural progenitor cells indicated substantial changes in neuronal gene expression. These data indicate that genome-wide significant associations with ASD implicate long noncoding RNAs. Because long noncoding RNAs are more abundant in human brain than protein-coding RNAs, this class of molecules is likely to contribute to ASD risk.

9.
Haematologica ; 104(12): 2361-2371, 2019 12.
Article in English | MEDLINE | ID: mdl-31004025

ABSTRACT

As high fetal hemoglobin levels ameliorate the underlying pathophysiological defects in sickle cell anemia and beta (ß)-thalassemia, understanding the mechanisms that enforce silencing of fetal hemoglobin postnatally offers the promise of effective molecular therapy. Depletion of the Nucleosome Remodeling and Deacetylase complex member MBD2 causes a 10-20-fold increase in γ-globin gene expression in adult ß-globin locus yeast artificial chromosome transgenic mice. To determine the effect of MBD2 depletion in human erythroid cells, genome editing technology was utilized to knockout MBD2 in Human Umbilical cord Derived Erythroid Progenitor-2 cells resulting in γ/γ+ß mRNA levels of approximately 50% and approximately 40% fetal hemoglobin by high performance liquid chromatography. In contrast, MBD3 knockout had no appreciable effect on γ-globin expression. Knockdown of MBD2 in primary adult erythroid cells consistently increased γ/γ+ß mRNA ratios by approximately 10-fold resulting in approximately 30-40% γ/γ+ß mRNA levels and a corresponding increase in γ-globin protein. MBD2 exerts its repressive effects through recruitment of the chromatin remodeler CHD4 via a coiled-coil domain, and the histone deacetylase core complex via an intrinsically disordered region. Enforced expression of wild-type MBD2 in MBD2 knockout cells caused a 5-fold decrease in γ-globin mRNA while neither the coiled-coil mutant nor the intrinsically disordered region mutant MBD2 proteins had an inhibitory effect. Co-immunoprecipitation assays showed that the coiled-coil and intrinsically disorder region mutations disrupt complex formation by dissociating the CHD4 and the histone deacetylase core complex components, respectively. These results establish the MBD2 Nucleosome Remodeling and Deacetylase complex as a major silencer of fetal hemoglobin in human erythroid cells and point to the coiled-coil and intrinsically disordered region of MBD2 as potential therapeutic targets.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , DNA-Binding Proteins/metabolism , Erythroid Cells/metabolism , Fetal Hemoglobin/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mutation , gamma-Globins/metabolism , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/genetics , Adult , Cells, Cultured , Chromatin Assembly and Disassembly , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Erythroid Cells/cytology , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/antagonists & inhibitors , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , ATP-Binding Cassette Sub-Family B Member 4
10.
Molecules ; 25(1)2019 Dec 30.
Article in English | MEDLINE | ID: mdl-31905878

ABSTRACT

Illustrated here is the development of a new class of antibiotic lead molecules targeted at Pseudomonas aeruginosa glutaredoxin (PaGRX). This lead was produced to (a) circumvent efflux-mediated resistance mechanisms via covalent inhibition while (b) taking advantage of species selectivity to target a fundamental metabolic pathway. This work involved four components: a novel workflow for generating protein specific fragment hits via independent nuclear magnetic resonance (NMR) measurements, NMR-based modeling of the target protein structure, NMR guided docking of hits, and synthetic modification of the fragment hit with a vinyl cysteine trap moiety, i.e., acrylamide warhead, to generate the chimeric lead. Reactivity of the top warhead-fragment lead suggests that the ortholog selectivity observed for a fragment hit can translate into a substantial kinetic advantage in the mature warhead lead, which bodes well for future work to identify potent, species specific drug molecules targeted against proteins heretofore deemed undruggable.


Subject(s)
Acrylamide/chemistry , Anti-Bacterial Agents/chemical synthesis , Glutaredoxins/antagonists & inhibitors , Lead/chemistry , Pseudomonas aeruginosa/enzymology , Small Molecule Libraries/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Glutaredoxins/chemistry , Humans , Kinetics , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Pseudomonas aeruginosa/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Species Specificity , Structure-Activity Relationship
11.
Physiol Genomics ; 50(8): 563-579, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29727589

ABSTRACT

Genomic sequencing has undergone massive expansion in the past 10 yr, from a rarely used research tool into an approach that has broad applications in a clinical setting. From rare disease to cancer, genomics is transforming our knowledge of biology. The transition from targeted gene sequencing, to whole exome sequencing, to whole genome sequencing has only been made possible due to rapid advancements in technologies and informatics that have plummeted the cost per base of DNA sequencing and analysis. The tools of genomics have resolved the etiology of disease for previously undiagnosable conditions, identified cancer driver gene variants, and have impacted the understanding of pathophysiology for many diseases. However, this expansion of use has also highlighted research's current voids in knowledge. The lack of precise animal models for gene-to-function association, lack of tools for analysis of genomic structural changes, skew in populations used for genetic studies, publication biases, and the "Unknown Proteome" all contribute to voids needing filled for genomics to work in a fast-paced clinical setting. The future will hold the tools to fill in these voids, with new data sets and the continual development of new technologies allowing for expansion of genomic medicine, ushering in the days to come for precision medicine. In this review we highlight these and other points in hopes of advancing and guiding precision medicine into the future for optimal success.


Subject(s)
Disease/genetics , Exome Sequencing/methods , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Neoplasms/genetics , Sequence Analysis, DNA/methods , Animals , Computational Biology/methods , Computational Biology/trends , Forecasting , Genomics/trends , High-Throughput Nucleotide Sequencing/trends , Humans , Precision Medicine/methods , Precision Medicine/trends , Sequence Analysis, DNA/trends , Exome Sequencing/trends
12.
Nucleic Acids Res ; 45(15): 9164-9177, 2017 Sep 06.
Article in English | MEDLINE | ID: mdl-28637186

ABSTRACT

The methylcytosine-binding domain 2 (MBD2) protein recruits the nucleosome remodeling and deacetylase complex (NuRD) to methylated DNA to modify chromatin and regulate transcription. Importantly, MBD2 functions within CpG islands that contain 100s to 1000s of potential binding sites. Since NuRD physically rearranges nucleosomes, the dynamic mobility of this complex is directly related to function. In these studies, we use NMR and single-molecule atomic force microscopy and fluorescence imaging to study DNA binding dynamics of MBD2. Single-molecule fluorescence tracking on DNA tightropes containing regions with CpG-rich and CpG-free regions reveals that MBD2 carries out unbiased 1D diffusion on CpG-rich DNA but subdiffusion on CpG-free DNA. In contrast, the protein stably and statically binds to methylated CpG (mCpG) regions. The intrinsically disordered region (IDR) on MBD2 both reduces exchange between mCpG sites along the DNA as well as the dissociation from DNA, acting like an anchor that restricts the dynamic mobility of the MBD domain. Unexpectedly, MBD2 binding to methylated CpGs induces DNA bending that is augmented by the IDR region of the protein. These results suggest that MBD2 targets NuRD to unmethylated or methylated CpG islands where its distinct dynamic binding modes help maintain open or closed chromatin, respectively.


Subject(s)
5-Methylcytosine/chemistry , CpG Islands , DNA-Binding Proteins/chemistry , DNA/chemistry , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Nucleosomes/metabolism , 5-Methylcytosine/metabolism , Animals , Binding Sites , Chickens , Cloning, Molecular , DNA/metabolism , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Fluorescence Polarization , Gene Expression , Humans , Magnetic Resonance Spectroscopy , Mi-2 Nucleosome Remodeling and Deacetylase Complex/ultrastructure , Nucleic Acid Conformation , Nucleosomes/ultrastructure , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Single Molecule Imaging
13.
J Mol Biol ; 429(10): 1581-1594, 2017 05 19.
Article in English | MEDLINE | ID: mdl-28450074

ABSTRACT

The Rett-syndrome-associated methyl-CpG-binding protein 2 (MeCP2) selectively binds methylated DNA to regulate transcription during the development of mature neurons. Like other members of the methyl-CpG-binding domain (MBD) family, MeCP2 functions through the recognition of symmetrical 5-methylcytosines in CpG (mCG) dinucleotides. Advances in base-level resolution epigenetic mapping techniques have revealed, however, that MeCP2 can bind asymmetrically methylated and hydroxymethylated CpA dinucleotides and that this alternative binding selectivity modifies gene expression in the developing mammalian brain. The structural determinants of binding to methylated CpA (mCA) and hydroxymethylated DNA have not been previously investigated. Here, we employ isothermal titration calorimetry and NMR spectroscopy to characterize MeCP2 binding to methylated and hydroxymethylated mCG and mCA DNA, examine the effects of Rett-syndrome-associated missense mutations, and make comparisons to the related and evolutionarily most ancient protein, MBD2. These analyses reveal that MeCP2 binds mCA with high affinity in a strand-specific and orientation-dependent manner. In contrast, MBD2 does not show high affinity or methyl-specific binding to mCA. The Rett-associated missense mutations (T158M, R106W, and P101S) destabilize the MeCP2 MBD and disrupt the recognition of mCG and mCA equally. Finally, hydroxymethylation of a high-affinity mCA site does not alter the binding properties, whereas hemi-hydroxylation of the equivalent cytosine in an mCG site decreases affinity and specificity. Based on these findings, we suggest that MeCP2 recognition of methylated/hydroxymethylated CpA dinucleotides functions as an epigenetic switch redistributing MeCP2 among mCG and mCA loci.


Subject(s)
DNA/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Calorimetry , DNA Mutational Analysis , Humans , Magnetic Resonance Spectroscopy , Methyl-CpG-Binding Protein 2/genetics , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Protein Binding
14.
Sci Rep ; 7: 40674, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28094816

ABSTRACT

DNA cytosine methylation and methyl-cytosine binding domain (MBD) containing proteins are found throughout all vertebrate species studied to date. However, both the presence of DNA methylation and pattern of methylation varies among invertebrate species. Invertebrates generally have only a single MBD protein, MBD2/3, that does not always contain appropriate residues for selectively binding methylated DNA. Therefore, we sought to determine whether sponges, one of the most ancient extant metazoan lineages, possess an MBD2/3 capable of recognizing methylated DNA and recruiting the associated nucleosome remodeling and deacetylase (NuRD) complex. We find that Ephydatia muelleri has genes for each of the NuRD core components including an EmMBD2/3 that selectively binds methylated DNA. NMR analyses reveal a remarkably conserved binding mode, showing almost identical chemical shift changes between binding to methylated and unmethylated CpG dinucleotides. In addition, we find that EmMBD2/3 and EmGATAD2A/B proteins form a coiled-coil interaction known to be critical for the formation of NuRD. Finally, we show that knockdown of EmMBD2/3 expression disrupts normal cellular architecture and development of E. muelleri. These data support a model in which the MBD2/3 methylation-dependent functional role emerged with the earliest multicellular organisms and has been maintained to varying degrees across animal evolution.


Subject(s)
Chromatin Assembly and Disassembly , DNA Methylation , Porifera/genetics , Amino Acid Sequence , Animals , DNA/chemistry , DNA/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Knockdown Techniques , Humans , Models, Molecular , Nucleic Acid Conformation , Phenotype , Porifera/metabolism , Protein Conformation
15.
Molecules ; 21(7)2016 Jul 16.
Article in English | MEDLINE | ID: mdl-27438815

ABSTRACT

Selective hits for the glutaredoxin ortholog of Brucella melitensis are determined using STD NMR and verified by trNOE and (15)N-HSQC titration. The most promising hit, RK207, was docked into the target molecule using a scoring function to compare simulated poses to experimental data. After elucidating possible poses, the hit was further optimized into the lead compound by extension with an electrophilic acrylamide warhead. We believe that focusing on selectivity in this early stage of drug discovery will limit cross-reactivity that might occur with the human ortholog as the lead compound is optimized. Kinetics studies revealed that lead compound 5 modified with an ester group results in higher reactivity than an acrylamide control; however, after modification this compound shows little selectivity for bacterial protein versus the human ortholog. In contrast, hydrolysis of compound 5 to the acid form results in a decrease in the activity of the compound. Together these results suggest that more optimization is warranted for this simple chemical scaffold, and opens the door for discovery of drugs targeted against glutaredoxin proteins-a heretofore untapped reservoir for antibiotic agents.


Subject(s)
Drug Discovery , Ligands , Molecular Docking Simulation , Proteins/chemistry , Binding Sites , Drug Discovery/methods , Drug Evaluation, Preclinical , Humans , Magnetic Resonance Spectroscopy , Molecular Conformation , Molecular Dynamics Simulation , Protein Binding , Proteins/antagonists & inhibitors , Small Molecule Libraries , Structure-Activity Relationship
16.
Sci Rep ; 6: 18278, 2016 Jan 04.
Article in English | MEDLINE | ID: mdl-26726850

ABSTRACT

The product of the meiosis-expressed gene 1 (MEIG1) is found in the cell bodies of spermatocytes and recruited to the manchette, a structure unique to elongating spermatids, by Parkin co-regulated gene (PACRG). This complex is essential for targeting cargo to the manchette during sperm flagellum assembly. Here we show that MEIG1 adopts a unique fold that provides a large surface for interacting with other proteins. We mutated 12 exposed and conserved amino acids and show that four of these mutations (W50A, K57E, F66A, Y68A) dramatically reduce binding to PACRG. These four amino acids form a contiguous hydrophobic patch on one end of the protein. Furthermore, each of these four mutations diminishes the ability of MEIG1 to stabilize PACRG when expressed in bacteria. Together these studies establish the unique structure and key interaction surface of MEIG1 and provide a framework to explore how MEIG1 recruits proteins to build the sperm tail.


Subject(s)
Cell Cycle Proteins/chemistry , Models, Molecular , Nuclear Proteins/chemistry , Phosphoproteins/chemistry , Protein Conformation , Proteins/chemistry , Amino Acid Sequence , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Mice , Microfilament Proteins , Molecular Chaperones , Molecular Sequence Data , Mutation , Nuclear Magnetic Resonance, Biomolecular , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Binding , Protein Folding , Protein Stability , Proteins/genetics , Proteins/metabolism , Recombinant Proteins , Solvents , Structure-Activity Relationship
17.
Gen Comp Endocrinol ; 215: 106-16, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25260253

ABSTRACT

The details of protein pathways at a structural level provides a bridge between genetics/molecular biology and physiology. The renin-angiotensin system is involved in many physiological pathways with informative structural details in multiple components. Few studies have been performed assessing structural knowledge across the system. This assessment allows use of bioinformatics tools to fill in missing structural voids. In this paper we detail known structures of the renin-angiotensin system and use computational approaches to estimate and model components that do not have their protein structures defined. With the subsequent large library of protein structures, we then created a species specific protein library for human, mouse, rat, bovine, zebrafish, and chicken for the system. The rat structural system allowed for rapid screening of genetic variants from 51 commonly used rat strains, identifying amino acid variants in angiotensinogen, ACE2, and AT1b that are in contact positions with other macromolecules. We believe the structural map will be of value for other researchers to understand their experimental data in the context of an environment for multiple proteins, providing pdb files of proteins for the renin-angiotensin system in six species. With detailed structural descriptions of each protein, it is easier to assess a species for use in translating human diseases with animal models. Additionally, as whole genome sequencing continues to decrease in cost, tools such as molecular modeling will gain use as an initial step in designing efficient hypothesis driven research, addressing potential functional outcomes of genetic variants with precompiled protein libraries aiding in rapid characterizations.


Subject(s)
Angiotensinogen/chemistry , Biological Evolution , Computational Biology , Models, Molecular , Renin-Angiotensin System , Renin/chemistry , Amino Acid Sequence , Angiotensinogen/metabolism , Animals , Cattle , Chickens , Humans , Mice , Molecular Sequence Data , Protein Conformation , Rats , Renin/metabolism , Sequence Homology, Amino Acid , Species Specificity , Zebrafish
18.
J Mol Biol ; 426(8): 1753-65, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24486611

ABSTRACT

Steroid receptor activator RNA protein (SRA1p) is the translation product of the bi-functional long non-coding RNA steroid receptor activator RNA 1 (SRA1) that is part of the steroid receptor coactivator-1 acetyltransferase complex and is indicated to be an epigenetic regulatory component. Previously, the SRA1p protein was suggested to contain an RNA recognition motif (RRM) domain. We have determined the solution structure of the C-terminal domain of human SRA1p by NMR spectroscopy. Our structure along with sequence comparisons among SRA1p orthologs and against authentic RRM proteins indicates that it is not an RRM domain but rather an all-helical protein with a fold more similar to the PRP18 splicing factor. NMR spectroscopy on the full SRA1p protein suggests that this structure is relevant to the native full-length context. Furthermore, molecular modeling indicates that this fold is well conserved among vertebrates. Amino acid variations in this protein seen across sequenced human genomes, including those in tumor cells, indicate that mutations that disrupt the fold occur vary rarely and highlight that its function is well conserved. SRA1p had previously been suggested to bind to the SRA1 RNA, but NMR spectra of SRA1p in the presence of its 80-nt RNA target suggest otherwise and indicate that this protein must be part of a multi-protein complex in order to recognize its proposed RNA recognition element.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Ribonucleoprotein, U5 Small Nuclear/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Amino Acid Sequence , Binding Sites , Carrier Proteins/genetics , Epigenesis, Genetic , Humans , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Nucleic Acid Conformation , Phylogeny , Protein Folding , Protein Interaction Domains and Motifs , RNA, Long Noncoding/chemistry , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribonucleoprotein, U5 Small Nuclear/genetics , Ribonucleoproteins/chemistry , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Sequence Homology, Amino Acid , Structural Homology, Protein
19.
Acta Biomater ; 10(2): 751-60, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24120846

ABSTRACT

Protein-polymer interactions are of great interest in a wide range of scientific and technological applications. Neutral poly(ethylene glycol) (PEG) and zwitterionic poly(sulfobetaine methacrylate) (pSBMA) are two well-known nonfouling materials that exhibit strong surface resistance to proteins. However, it still remains unclear or unexplored how PEG and pSBMA interact with proteins in solution. In this work, we examine the interactions between two model proteins (bovine serum albumin and lysozyme) and two typical antifouling polymers of PEG and pSBMA in aqueous solution using fluorescence spectroscopy, atomic force microscopy and nuclear magnetic resonance. The effect of protein:polymer mass ratios on the interactions is also examined. Collective data clearly demonstrate the existence of weak hydrophobic interactions between PEG and proteins, while there are no detectable interactions between pSBMA and proteins. The elimination of protein interaction with pSBMA could be due to an enhanced surface hydration of zwitterionic groups in pSBMA. New evidence is given to demonstrate the interactions between PEG and proteins, which are often neglected in the literature because the PEG-protein interactions are weak and reversible, as well as the structural change caused by hydrophobic interaction. This work provides a better fundamental understanding of the intrinsic structure-activity relationship of polymers underlying polymer-protein interactions, which are important for designing new biomaterials for biosensor, medical diagnostics and drug delivery applications.


Subject(s)
Biofouling , Methacrylates/metabolism , Muramidase/metabolism , Polyethylene Glycols/metabolism , Serum Albumin, Bovine/metabolism , Animals , Cattle , Fluorescence , Ions , Magnetic Resonance Spectroscopy , Microscopy, Atomic Force , Molecular Weight , Protein Binding
20.
J Inorg Biochem ; 128: 11-6, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23911566

ABSTRACT

Multiple biophysical methods demonstrate that silver effectively metallates Pseudomonas aeruginosa apo-azurin in solution. X-ray crystallography of the silver-modified protein reveals that silver binds to azurin at the traditional copper mediated active site with nearly identical geometry. Cyclic voltammetry indicates that the silver adduct is redox inert. Our results suggest that a potential mechanism for the microbial toxicity of silver is the deactivation of copper oxidoreductases by the effective binding and structural mimicry by silver without the corresponding function.


Subject(s)
Azurin/chemistry , Oxidoreductases/chemistry , Pseudomonas aeruginosa/enzymology , Silver/chemistry , Azurin/antagonists & inhibitors , Azurin/metabolism , Copper/chemistry , Copper/metabolism , Crystallography, X-Ray , Enzyme Activation/drug effects , Magnetic Resonance Spectroscopy , Models, Molecular , Oxidation-Reduction/drug effects , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/metabolism , Protein Conformation , Protein Structure, Tertiary , Silver/metabolism , Silver/pharmacology , Spectrometry, Mass, Electrospray Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...