Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 8 de 8
1.
Commun Biol ; 3(1): 363, 2020 07 09.
Article En | MEDLINE | ID: mdl-32647372

Histone H3.3 mutations are a hallmark of pediatric gliomas, but their core oncogenic mechanisms are not well-defined. To identify major effectors, we used CRISPR-Cas9 to introduce H3.3K27M and G34R mutations into previously H3.3-wildtype brain cells, while in parallel reverting the mutations in glioma cells back to wildtype. ChIP-seq analysis broadly linked K27M to altered H3K27me3 activity including within super-enhancers, which exhibited perturbed transcriptional function. This was largely independent of H3.3 DNA binding. The K27M and G34R mutations induced several of the same pathways suggesting key shared oncogenic mechanisms including activation of neurogenesis and NOTCH pathway genes. H3.3 mutant gliomas are also particularly sensitive to NOTCH pathway gene knockdown and drug inhibition, reducing their viability in culture. Reciprocal editing of cells generally produced reciprocal effects on tumorgenicity in xenograft assays. Overall, our findings define common and distinct K27M and G34R oncogenic mechanisms, including potentially targetable pathways.


Biomarkers, Tumor/metabolism , Gene Editing , Gene Expression Regulation, Neoplastic , Glioma/pathology , Histones/genetics , Mutation , Receptors, Notch/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Proliferation , Child , Female , Glioma/genetics , Glioma/metabolism , Glycine/chemistry , Glycine/genetics , Histones/chemistry , Humans , Lysine/chemistry , Lysine/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Notch/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Nature ; 584(7819): 125-129, 2020 08.
Article En | MEDLINE | ID: mdl-32528175

The D2 dopamine receptor (DRD2) is a therapeutic target for Parkinson's disease1 and antipsychotic drugs2. DRD2 is activated by the endogenous neurotransmitter dopamine and synthetic agonist drugs such as bromocriptine3, leading to stimulation of Gi and inhibition of adenylyl cyclase. Here we used cryo-electron microscopy to elucidate the structure of an agonist-bound activated DRD2-Gi complex reconstituted into a phospholipid membrane. The extracellular ligand-binding site of DRD2 is remodelled in response to agonist binding, with conformational changes in extracellular loop 2, transmembrane domain 5 (TM5), TM6 and TM7, propagating to opening of the intracellular Gi-binding site. The DRD2-Gi structure represents, to our knowledge, the first experimental model of a G-protein-coupled receptor-G-protein complex embedded in a phospholipid bilayer, which serves as a benchmark to validate the interactions seen in previous detergent-bound structures. The structure also reveals interactions that are unique to the membrane-embedded complex, including helix 8 burial in the inner leaflet, ordered lysine and arginine side chains in the membrane interfacial regions, and lipid anchoring of the G protein in the membrane. Our model of the activated DRD2 will help to inform the design of subtype-selective DRD2 ligands for multiple human central nervous system disorders.


Cryoelectron Microscopy , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/ultrastructure , Membrane Lipids/metabolism , Membranes, Artificial , Receptors, Dopamine D2/chemistry , Receptors, Dopamine D2/ultrastructure , Bromocriptine/chemistry , Bromocriptine/metabolism , Dopamine/chemistry , Dopamine/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Membrane Lipids/chemistry , Models, Molecular , Protein Conformation , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/metabolism , Signal Transduction
3.
Nat Chem Biol ; 16(1): 77-86, 2020 01.
Article En | MEDLINE | ID: mdl-31792443

Membrane receptors sense and transduce extracellular stimuli into intracellular signaling responses but the molecular underpinnings remain poorly understood. We report a computational approach for designing protein allosteric signaling functions. By combining molecular dynamics simulations and design calculations, the method engineers amino-acid 'microswitches' at allosteric sites that modulate receptor stability or long-range coupling, to reprogram specific signaling properties. We designed 36 dopamine D2 receptor variants, whose constitutive and ligand-induced signaling agreed well with our predictions, repurposed the D2 receptor into a serotonin biosensor and predicted the signaling effects of more than 100 known G-protein-coupled receptor (GPCR) mutations. Our results reveal the existence of distinct classes of allosteric microswitches and pathways that define an unforeseen molecular mechanism of regulation and evolution of GPCR signaling. Our approach enables the rational design of allosteric receptors with enhanced stability and function to facilitate structural characterization, and reprogram cellular signaling in synthetic biology and cell engineering applications.


Protein Engineering , Receptors, Dopamine D2/chemistry , Receptors, Dopamine D2/genetics , Signal Transduction , Allosteric Regulation/drug effects , Allosteric Site , Amino Acid Motifs , Biosensing Techniques , Computer Simulation , DNA Mutational Analysis , HEK293 Cells , Humans , Inhibitory Concentration 50 , Kinetics , Ligands , Molecular Dynamics Simulation , Mutagenesis , Serotonin/chemistry , Software
4.
Nat Chem Biol ; 13(7): 715-723, 2017 Jul.
Article En | MEDLINE | ID: mdl-28459439

Accurate modeling and design of protein-ligand interactions have broad applications in cell biology, synthetic biology and drug discovery but remain challenging without experimental protein structures. Here we developed an integrated protein-homology-modeling, ligand-docking protein-design approach that reconstructs protein-ligand binding sites from homolog protein structures in the presence of protein-bound ligand poses to capture conformational selection and induced-fit modes of ligand binding. In structure modeling tests, we blindly predicted, with near-atomic accuracy, ligand conformations bound to G-protein-coupled receptors (GPCRs) that have rarely been identified using traditional approaches. We also quantitatively predicted the binding selectivity of diverse ligands to structurally uncharacterized GPCRs. We then applied this technique to design functional human dopamine receptors with novel ligand-binding selectivity. Most blindly predicted ligand-binding specificities closely agreed with experimental validations. Our method should prove useful in ligand discovery approaches and in reprogramming the ligand-binding profile of membrane receptors that remain difficult to crystallize.


Computer-Aided Design , Ligands , Receptors, Dopamine/chemistry , Receptors, Dopamine/metabolism , Binding Sites , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Substrate Specificity
5.
Regen Med ; 11(8): 801-816, 2016 12.
Article En | MEDLINE | ID: mdl-27905217

The goal of editing the genomes of stem cells to generate model organisms and cell lines for genetic and biological studies has been pursued for decades. There is also exciting potential for future clinical impact in humans. While recent, rapid advances in targeted nuclease technologies have led to unprecedented accessibility and ease of gene editing, biology has benefited from past directed gene modification via homologous recombination, gene traps and other transgenic methodologies. Here we review the history of genome editing in stem cells (including via zinc finger nucleases, transcription activator-like effector nucleases and CRISPR-Cas9), discuss recent developments leading to the implementation of stem cell gene therapies in clinical trials and consider the prospects for future advances in this rapidly evolving field.


Clustered Regularly Interspaced Short Palindromic Repeats , Endonucleases/genetics , Gene Editing , Genetic Engineering/methods , Genome, Human , Stem Cells/metabolism , Animals , Humans , Stem Cells/cytology
6.
PLoS Comput Biol ; 10(5): e1003636, 2014 May.
Article En | MEDLINE | ID: mdl-24854015

Eukaryotic transmembrane helical (TMH) proteins perform a wide diversity of critical cellular functions, but remain structurally largely uncharacterized and their high-resolution structure prediction is currently hindered by the lack of close structural homologues. To address this problem, we present a novel and generic method for accurately modeling large TMH protein structures from distant homologues exhibiting distinct loop and TMH conformations. Models of the adenosine A2AR and chemokine CXCR4 receptors were first ranked in GPCR-DOCK blind prediction contests in the receptor structure accuracy category. In a benchmark of 50 TMH protein homolog pairs of diverse topology (from 5 to 12 TMHs), size (from 183 to 420 residues) and sequence identity (from 15% to 70%), the method improves most starting templates, and achieves near-atomic accuracy prediction of membrane-embedded regions. Unlike starting templates, the models are of suitable quality for computer-based protein engineering: redesigned models and redesigned X-ray structures exhibit very similar native interactions. The method should prove useful for the atom-level modeling and design of a large fraction of structurally uncharacterized TMH proteins from a wide range of structural homologues.


Models, Chemical , Molecular Docking Simulation/methods , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/ultrastructure , Receptors, CXCR4/chemistry , Receptors, CXCR4/ultrastructure , Amino Acid Sequence , Computer Simulation , Membrane Proteins/chemistry , Membrane Proteins/ultrastructure , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid
7.
Proc Natl Acad Sci U S A ; 109(33): 13284-9, 2012 Aug 14.
Article En | MEDLINE | ID: mdl-22847407

A wide range of membrane receptors signal through conformational changes, and the resulting protein conformational flexibility often hinders their structural studies. Because the determinants of membrane receptor conformational stability are still poorly understood, identifying a minimal set of perturbations stabilizing a membrane protein in a given conformation remains a major challenge in membrane protein structure determination. We present a novel approach integrating bioinformatics, computational design and experimental techniques that identifies and stabilizes metastable receptor regions. When applied to the beta1-adrenergic receptor, the method generated 13 novel receptor variants stabilized in the intended inactive state among which two exhibit an apparent thermostability higher than WT and M23 (a receptor variant previously stabilized by extensive scanning mutagenesis) by more than 30 °C and 11 °C, respectively. Targeted regions involve nonconserved unsatisfied polar residues or exhibit significant packing defects, features found in all class A G protein-coupled receptor structures. These findings suggest that natural G protein-coupled receptor sequences have evolved to be conformationally metastable through the design of suboptimal polar and van der Waals tertiary interactions. Given sufficiently accurate structural models, our approach should prove useful for designing stabilized variants of many uncharacterized membrane receptors.


Evolution, Molecular , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Amino Acid Motifs , Amino Acid Substitution/genetics , Amino Acids/chemistry , Conserved Sequence , Models, Molecular , Mutant Proteins/chemistry , Mutation/genetics , Protein Conformation , Protein Stability , Receptors, Adrenergic, beta-1/chemistry , Signal Transduction , Surface Properties , Thermodynamics
8.
Clin Cancer Res ; 17(8): 2373-84, 2011 Apr 15.
Article En | MEDLINE | ID: mdl-21372221

PURPOSE: This study evaluates the effect of dual PI3K and mTOR inhibition using NVP-BEZ235 in preclinical models of ovarian cancer as a potential novel therapeutic strategy. EXPERIMENTAL DESIGN: Inhibition of PI3K/Akt/mTOR signaling by NVP-BEZ235 was demonstrated by immunoblotting. The effect on cell proliferation was assessed in 18 ovarian cancer cell lines, including four pairs of syngeneic cisplatin-sensitive and cisplatin-resistant cell lines. The in vivo effects of NVP-BEZ235 on established tumor growth were evaluated using an immunocompetent, transgenic murine ovarian cancer model (LSL-K-ras(G12D/+)Pten(loxP/loxP)). RESULTS: NVP-BEZ235 decreased cell proliferation in all ovarian cancer cell lines assayed and sensitized cisplatin-resistant cells to the cytotoxic effects of cisplatin. Cell lines with PI3K-activating mutations or Pten deletions were significantly more sensitive to the effect of NVP-BEZ235 than cell lines without these mutations (P < 0.05). A statistically significant correlation was found between relative levels of p4E-BP1 and the IC(50) for NVP-BEZ235. In LSL-K-ras(G12D/+)Pten(loxP/loxP) mice with established intraperitoneal tumor disease, oral administration of NVP-BEZ235 decreased pAkt, p4E-BP1 and Ki67 in tumor tissue, and resulted in significantly longer survival compared to control animals (P < 0.05). NVP-BEZ235 also induced cell cycle arrest, caspase 3 activity, and reduced cell migration. CONCLUSIONS: Targeting PI3K and mTOR simultaneously using NVP-BEZ235 effectively inhibits ovarian cancer cell growth even in the presence of platinum resistance and prolongs survival of mice with intra-abdominal ovarian tumor disease. We propose that dual PI3K and mTOR inhibition using NVP-BEZ235 may be an effective novel therapeutic approach in patients with ovarian cancer.


Imidazoles/therapeutic use , Ovarian Neoplasms/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Quinolines/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cisplatin/pharmacology , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Enzyme Activation/drug effects , Female , Humans , Immunoblotting , Mice , Mice, Transgenic , Mutation , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
...