Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Nature ; 608(7921): 174-180, 2022 08.
Article in English | MEDLINE | ID: mdl-35732739

ABSTRACT

Heart failure encompasses a heterogeneous set of clinical features that converge on impaired cardiac contractile function1,2 and presents a growing public health concern. Previous work has highlighted changes in both transcription and protein expression in failing hearts3,4, but may overlook molecular changes in less prevalent cell types. Here we identify extensive molecular alterations in failing hearts at single-cell resolution by performing single-nucleus RNA sequencing of nearly 600,000 nuclei in left ventricle samples from 11 hearts with dilated cardiomyopathy and 15 hearts with hypertrophic cardiomyopathy as well as 16 non-failing hearts. The transcriptional profiles of dilated or hypertrophic cardiomyopathy hearts broadly converged at the tissue and cell-type level. Further, a subset of hearts from patients with cardiomyopathy harbour a unique population of activated fibroblasts that is almost entirely absent from non-failing samples. We performed a CRISPR-knockout screen in primary human cardiac fibroblasts to evaluate this fibrotic cell state transition; knockout of genes associated with fibroblast transition resulted in a reduction of myofibroblast cell-state transition upon TGFß1 stimulation for a subset of genes. Our results provide insights into the transcriptional diversity of the human heart in health and disease as well as new potential therapeutic targets and biomarkers for heart failure.


Subject(s)
Cardiomyopathy, Dilated , Cardiomyopathy, Hypertrophic , Cell Nucleus , Gene Expression Profiling , Heart Failure , Single-Cell Analysis , CRISPR-Cas Systems , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Case-Control Studies , Cell Nucleus/genetics , Cells, Cultured , Gene Knockout Techniques , Heart Failure/genetics , Heart Failure/pathology , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Myocardium/metabolism , Myocardium/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , RNA-Seq , Transcription, Genetic , Transforming Growth Factor beta1
2.
Bioinformatics ; 38(6): 1756-1760, 2022 03 04.
Article in English | MEDLINE | ID: mdl-34935911

ABSTRACT

SUMMARY: Accurately identifying cell types is a critical step in single-cell sequencing analyses. Here, we present marker-based automatic cell-type annotation (MACA), a new tool for annotating single-cell transcriptomics datasets. We developed MACA by testing four cell-type scoring methods with two public cell-marker databases as reference in six single-cell studies. MACA compares favorably to four existing marker-based cell-type annotation methods in terms of accuracy and speed. We show that MACA can annotate a large single-nuclei RNA-seq study in minutes on human hearts with ∼290K cells. MACA scales easily to large datasets and can broadly help experts to annotate cell types in single-cell transcriptomics datasets, and we envision MACA provides a new opportunity for integration and standardization of cell-type annotation across multiple datasets. AVAILABILITY AND IMPLEMENTATION: MACA is written in python and released under GNU General Public License v3.0. The source code is available at https://github.com/ImXman/MACA. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Lepidium , Software , Humans , RNA-Seq , Gene Expression Profiling , Databases, Factual , Single-Cell Analysis
3.
Proc Natl Acad Sci U S A ; 116(7): 2551-2560, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30683722

ABSTRACT

Since the late 1980s, mutations in the RAS genes have been recognized as major oncogenes with a high occurrence rate in human cancers. Such mutations reduce the ability of the small GTPase RAS to hydrolyze GTP, keeping this molecular switch in a constitutively active GTP-bound form that drives, unchecked, oncogenic downstream signaling. One strategy to reduce the levels of active RAS is to target guanine nucleotide exchange factors, which allow RAS to cycle from the inactive GDP-bound state to the active GTP-bound form. Here, we describe the identification of potent and cell-active small-molecule inhibitors which efficiently disrupt the interaction between KRAS and its exchange factor SOS1, a mode of action confirmed by a series of biophysical techniques. The binding sites, mode of action, and selectivity were elucidated using crystal structures of KRASG12C-SOS1, SOS1, and SOS2. By preventing formation of the KRAS-SOS1 complex, these inhibitors block reloading of KRAS with GTP, leading to antiproliferative activity. The final compound 23 (BAY-293) selectively inhibits the KRAS-SOS1 interaction with an IC50 of 21 nM and is a valuable chemical probe for future investigations.


Subject(s)
Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , SOS1 Protein/antagonists & inhibitors , Cell Line , Crystallography, X-Ray , Drug Discovery , Fluorescence Resonance Energy Transfer , High-Throughput Screening Assays , Humans , Protein Binding , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/metabolism , SOS1 Protein/chemistry , SOS1 Protein/metabolism , Signal Transduction
4.
Circulation ; 142(5): 466-482, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32403949

ABSTRACT

BACKGROUND: The human heart requires a complex ensemble of specialized cell types to perform its essential function. A greater knowledge of the intricate cellular milieu of the heart is critical to increase our understanding of cardiac homeostasis and pathology. As recent advances in low-input RNA sequencing have allowed definitions of cellular transcriptomes at single-cell resolution at scale, we have applied these approaches to assess the cellular and transcriptional diversity of the nonfailing human heart. METHODS: Microfluidic encapsulation and barcoding was used to perform single nuclear RNA sequencing with samples from 7 human donors, selected for their absence of overt cardiac disease. Individual nuclear transcriptomes were then clustered based on transcriptional profiles of highly variable genes. These clusters were used as the basis for between-chamber and between-sex differential gene expression analyses and intersection with genetic and pharmacologic data. RESULTS: We sequenced the transcriptomes of 287 269 single cardiac nuclei, revealing 9 major cell types and 20 subclusters of cell types within the human heart. Cellular subclasses include 2 distinct groups of resident macrophages, 4 endothelial subtypes, and 2 fibroblast subsets. Comparisons of cellular transcriptomes by cardiac chamber or sex reveal diversity not only in cardiomyocyte transcriptional programs but also in subtypes involved in extracellular matrix remodeling and vascularization. Using genetic association data, we identified strong enrichment for the role of cell subtypes in cardiac traits and diseases. Intersection of our data set with genes on cardiac clinical testing panels and the druggable genome reveals striking patterns of cellular specificity. CONCLUSIONS: Using large-scale single nuclei RNA sequencing, we defined the transcriptional and cellular diversity in the normal human heart. Our identification of discrete cell subtypes and differentially expressed genes within the heart will ultimately facilitate the development of new therapeutics for cardiovascular diseases.


Subject(s)
Myocardium/cytology , Transcription, Genetic , Adipocytes/metabolism , Adult , Aged , Cardiovascular Agents/pharmacology , Cardiovascular Agents/therapeutic use , Endothelial Cells/classification , Endothelial Cells/metabolism , Fibroblasts/classification , Fibroblasts/metabolism , Gene Ontology , Heart/innervation , Heart Atria/cytology , Heart Diseases/drug therapy , Heart Ventricles/cytology , Homeostasis , Humans , Lymphocyte Subsets/metabolism , Macrophages/classification , Macrophages/metabolism , Microfluidic Analytical Techniques , Middle Aged , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Smooth Muscle/metabolism , Pericytes/metabolism , RNA-Seq , Sex Characteristics , Single-Cell Analysis , Transcriptome
5.
J Biol Chem ; 291(21): 11252-67, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27030009

ABSTRACT

The spindle assembly checkpoint (SAC) is an essential safeguarding mechanism devised to ensure equal chromosome distribution in daughter cells upon mitosis. The proteins Bub3 and BubR1 are key components of the mitotic checkpoint complex, an essential part of the molecular machinery on which the SAC relies. In the present work we have performed a detailed functional and biochemical characterization of the interaction between human Bub3 and BubR1 in cells and in vitro Our results demonstrate that genetic knockdown of Bub3 abrogates the SAC, promotes apoptosis, and inhibits the proliferation of human cancer cells. We also show that the integrity of the human mitotic checkpoint complex depends on the specific recognition between BubR1 and Bub3, for which the BubR1 Gle2 binding sequence motif is essential. This 1:1 binding event is high affinity, enthalpy-driven and with slow dissociation kinetics. The affinity, kinetics, and thermodynamic parameters of the interaction are differentially modulated by small regions in the N and C termini of the Gle2 binding domain sequence, suggesting the existence of "hotspots" for this protein-protein interaction. Furthermore, we show that specific disruption of endogenous BubR1·Bub3 complexes in human cancer cells phenocopies the effects observed in gene targeting experiments. Our work enhances the current understanding of key members of the SAC and paves the road for the pursuit of novel targeted cancer therapies based on SAC inhibition.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , M Phase Cell Cycle Checkpoints/physiology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Spindle Apparatus/metabolism , Apoptosis , Cell Cycle Proteins/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation , Gene Knockdown Techniques , HeLa Cells , Humans , Kinetics , M Phase Cell Cycle Checkpoints/genetics , MCF-7 Cells , Models, Molecular , Poly-ADP-Ribose Binding Proteins , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spindle Apparatus/genetics , Thermodynamics
7.
Proc Natl Acad Sci U S A ; 108(37): 15264-9, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21876177

ABSTRACT

SNARE protein-driven secretion of neurotransmitters from synaptic vesicles is at the center of neuronal communication. In the absence of the cytosolic protein Munc18-1, synaptic secretion comes to a halt. Although it is believed that Munc18-1 orchestrates SNARE complexes, its mode of action is still a matter of debate. In particular, it has been challenging to clarify the role of a tight Munc18/syntaxin 1 complex, because this interaction interferes strongly with syntaxin's ability to form a SNARE complex. In this complex, two regions of syntaxin, the N-peptide and the remainder in closed conformation, bind to Munc18 simultaneously. Until now, this binary complex has been reported for neuronal tissues only, leading to the hypothesis that it might be a specialization of the neuronal secretion apparatus. Here we aimed, by comparing the core secretion machinery of the unicellular choanoflagellate Monosiga brevicollis with that of animals, to reconstruct the ancestral function of the Munc18/syntaxin1 complex. We found that the Munc18/syntaxin 1 complex from M. brevicollis is structurally and functionally highly similar to the vertebrate complex, suggesting that it constitutes a fundamental step in the reaction pathway toward SNARE assembly. We thus propose that the primordial secretion machinery of the common ancestor of choanoflagellates and animals has been co-opted for synaptic roles during the rise of animals.


Subject(s)
Choanoflagellata/metabolism , Neurosecretory Systems/metabolism , Choanoflagellata/cytology , Choanoflagellata/drug effects , Choanoflagellata/ultrastructure , Crystallography, X-Ray , Detergents/pharmacology , Munc18 Proteins/chemistry , Munc18 Proteins/metabolism , Neurosecretory Systems/drug effects , Neurosecretory Systems/ultrastructure , Phylogeny , Protein Binding/drug effects , Protein Structure, Secondary , SNARE Proteins/metabolism , Synapses/drug effects , Synapses/metabolism , Syntaxin 1/chemistry , Syntaxin 1/metabolism , Thermodynamics
8.
bioRxiv ; 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-38014050

ABSTRACT

Background: Despite the critical role of the cardiovascular system, our understanding of its cellular and transcriptional diversity remains limited. We therefore sought to characterize the cellular composition, phenotypes, molecular pathways, and communication networks between cell types at the tissue and sub-tissue level across the cardiovascular system of the healthy Wistar rat, an important model in preclinical cardiovascular research. We obtained high quality tissue samples under controlled conditions that reveal a level of cellular detail so far inaccessible in human studies. Methods and Results: We performed single nucleus RNA-sequencing in 78 samples in 10 distinct regions including the four chambers of the heart, ventricular septum, sinoatrial node, atrioventricular node, aorta, pulmonary artery, and pulmonary veins (PV), which produced an aggregate map of 505,835 nuclei. We identified 26 distinct cell types and additional subtypes, including a number of rare cell types such as PV cardiomyocytes and non-myelinating Schwann cells (NMSCs), and unique groups of vascular smooth muscle cells (VSMCs), endothelial cells (ECs) and fibroblasts (FBs), which gave rise to a detailed cell type distribution across tissues. We demonstrated differences in the cellular composition across different cardiac regions and tissue-specific differences in transcription for each cell type, highlighting the molecular diversity and complex tissue architecture of the cardiovascular system. Specifically, we observed great transcriptional heterogeneities among ECs and FBs. Importantly, several cell subtypes had a unique regional localization such as a subtype of VSMCs enriched in the large vasculature. We found the cellular makeup of PV tissue is closer to heart tissue than to the large arteries. We further explored the ligand-receptor repertoire across cell clusters and tissues, and observed tissue-enriched cellular communication networks, including heightened Nppa - Npr1/2/3 signaling in the sinoatrial node. Conclusions: Through a large single nucleus sequencing effort encompassing over 500,000 nuclei, we broadened our understanding of cellular transcription in the healthy cardiovascular system. The existence of tissue-restricted cellular phenotypes suggests regional regulation of cardiovascular physiology. The overall conservation in gene expression and molecular pathways across rat and human cell types, together with our detailed transcriptional characterization of each cell type, offers the potential to identify novel therapeutic targets and improve preclinical models of cardiovascular disease.

9.
Nat Genet ; 54(1): 40-51, 2022 01.
Article in English | MEDLINE | ID: mdl-34837083

ABSTRACT

Enlargement or aneurysm of the aorta predisposes to dissection, an important cause of sudden death. We trained a deep learning model to evaluate the dimensions of the ascending and descending thoracic aorta in 4.6 million cardiac magnetic resonance images from the UK Biobank. We then conducted genome-wide association studies in 39,688 individuals, identifying 82 loci associated with ascending and 47 with descending thoracic aortic diameter, of which 14 loci overlapped. Transcriptome-wide analyses, rare-variant burden tests and human aortic single nucleus RNA sequencing prioritized genes including SVIL, which was strongly associated with descending aortic diameter. A polygenic score for ascending aortic diameter was associated with thoracic aortic aneurysm in 385,621 UK Biobank participants (hazard ratio = 1.43 per s.d., confidence interval 1.32-1.54, P = 3.3 × 10-20). Our results illustrate the potential for rapidly defining quantitative traits with deep learning, an approach that can be broadly applied to biomedical images.


Subject(s)
Aorta, Thoracic/anatomy & histology , Deep Learning , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging , Adult , Aged , Aorta, Thoracic/pathology , Aortic Aneurysm/genetics , Aortic Aneurysm/pathology , Biological Variation, Population , Female , Genome-Wide Association Study , Humans , Male , Middle Aged , Quantitative Trait Loci , Transcriptome
10.
medRxiv ; 2020 Apr 14.
Article in English | MEDLINE | ID: mdl-32511660

ABSTRACT

Coronavirus disease 2019 (COVID-19) is a global pandemic caused by a novel severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). SARS-CoV-2 infection of host cells occurs predominantly via binding of the viral surface spike protein to the human angiotensin-converting enzyme 2 (ACE2) receptor. Hypertension and pre-existing cardiovascular disease are risk factors for morbidity from COVID-19, and it remains uncertain whether the use of angiotensin converting enzyme inhibitors (ACEi) or angiotensin receptor blockers (ARB) impacts infection and disease. Here, we aim to shed light on this question by assessing ACE2 expression in normal and diseased human myocardial samples profiled by bulk and single nucleus RNA-seq.

11.
Angew Chem Int Ed Engl ; 48(28): 5207-10, 2009.
Article in English | MEDLINE | ID: mdl-19499554

ABSTRACT

Water molecules doing time: Atomic-resolution crystal structures of the PPIase domain of cyclophilin G, alone and in complex with cyclosporin A, and together with MD simulations and calorimetry, reveal how trapped water molecules influence the thermodynamic profile of a protein-ligand interaction.


Subject(s)
Cyclophilins/chemistry , Water/chemistry , Crystallography, X-Ray , Cyclosporine/chemistry , Humans , Hydrogen Bonding , Ligands , Protein Structure, Tertiary , Thermodynamics
12.
Front Biosci (Landmark Ed) ; 22(1): 21-47, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27814600

ABSTRACT

The impact of target binding kinetics (BK) on the clinical performance of therapeutic agents is presently a topic of intense debate in drug discovery. While retrospective studies suggest that BK is a differentiating parameter in marketed medicines, it is yet unclear how this information could be used to prioritize drug candidates during lead optimization. Motivated by the question whether BK can be understood and rationally optimized, we review the most relevant literature in the field, with special focus on selected examples from our organization. First we discuss structure-kinetic relationships (SKR), and how they can be influenced by factors such as conformational changes, molecular flexibility, hydrogen bonds, hydrophobicity, water molecules and (reversible-) covalent bonds. We then introduce the methodologies currently used for the investigation of BK parameters, briefly commenting on their strengths, weaknesses and future trends. Finally, we present our current perspective on the integration of BK in the drug discovery process, aiming to stimulate further thoughts on this important subject.


Subject(s)
Drug Discovery/methods , Animals , Drug Discovery/trends , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Ligands , Molecular Conformation , Niacinamide/analogs & derivatives , Niacinamide/pharmacokinetics , Niacinamide/pharmacology , Oligopeptides/chemistry , Oligopeptides/metabolism , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/pharmacology , Protein Binding , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/chemistry , Sorafenib , Structure-Activity Relationship
13.
ACS Chem Biol ; 11(6): 1710-9, 2016 06 17.
Article in English | MEDLINE | ID: mdl-27090615

ABSTRACT

Roniciclib (BAY 1000394) is a type I pan-CDK (cyclin-dependent kinase) inhibitor which has revealed potent efficacy in xenograft cancer models. Here, we show that roniciclib displays prolonged residence times on CDK2 and CDK9, whereas residence times on other CDKs are transient, thus giving rise to a kinetic selectivity of roniciclib. Surprisingly, variation of the substituent at the 5-position of the pyrimidine scaffold results in changes of up to 3 orders of magnitude of the drug-target residence time. CDK2 X-ray cocrystal structures have revealed a DFG-loop adaption for the 5-(trifluoromethyl) substituent, while for hydrogen and bromo substituents the DFG loop remains in its characteristic type I inhibitor position. In tumor cells, the prolonged residence times of roniciclib on CDK2 and CDK9 are reflected in a sustained inhibitory effect on retinoblastoma protein (RB) phosphorylation, indicating that the target residence time on CDK2 may contribute to sustained target engagement and antitumor efficacy.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/pharmacokinetics , Sulfoxides/pharmacokinetics , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/chemistry , Aurora Kinase A/antagonists & inhibitors , HeLa Cells , Humans , Kinetics , MCF-7 Cells , Mice , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/chemistry , Pyrimidines/blood , Pyrimidines/chemistry , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Sulfonamides/pharmacokinetics , Sulfoxides/blood , Sulfoxides/chemistry
14.
J Med Chem ; 59(3): 1176-83, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26824386

ABSTRACT

Protein methyltransferases (PMTs) are a promising target class in oncology and other disease areas. They are composed of SET domain methyltransferases and structurally unrelated Rossman-fold enzymes that include protein arginine methyltransferases (PRMTs). In the absence of a well-defined medicinal chemistry tool-kit focused on PMTs, most current inhibitors were identified by screening large and diverse libraries of leadlike molecules. So far, no successful fragment-based approach was reported against this target class. Here, by deconstructing potent PRMT inhibitors, we find that chemical moieties occupying the substrate arginine-binding site can act as efficient fragment inhibitors. Screening a fragment library against PRMT6 produced numerous hits, including a 300 nM inhibitor (ligand efficiency of 0.56) that decreased global histone 3 arginine 2 methylation in cells, and can serve as a warhead for the development of PRMT chemical probes.


Subject(s)
Drug Discovery , Enzyme Inhibitors/pharmacology , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Repressor Proteins/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Structure-Activity Relationship
15.
Structure ; 21(3): 365-75, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23415559

ABSTRACT

NusG is a conserved regulatory protein interacting with RNA polymerase (RNAP) and other proteins to form multicomponent complexes that modulate transcription. The crystal structure of Thermotoga maritima NusG (TmNusG) shows a three-domain architecture, comprising well-conserved amino-terminal (NTD) and carboxy-terminal (CTD) domains with an additional, species-specific domain inserted into the NTD. NTD and CTD directly contact each other, occluding a surface of the NTD for binding to RNAP and a surface on the CTD interacting either with transcription termination factor Rho or transcription antitermination factor NusE. NMR spectroscopy confirmed the intramolecular NTD-CTD interaction up to the optimal growth temperature of Thermotoga maritima. The domain interaction involves a dynamic equilibrium between open and closed states and contributes significantly to the overall fold stability of the protein. Wild-type TmNusG and deletion variants could not replace endogenous Escherichia coli NusG, suggesting that the NTD-CTD interaction of TmNusG represents an autoinhibited state.


Subject(s)
Bacterial Proteins/chemistry , Escherichia coli Proteins/chemistry , Peptide Elongation Factors/chemistry , Thermotoga maritima/chemistry , Transcription Factors/chemistry , Bacterial Proteins/genetics , Binding Sites , Crystallography, X-Ray , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/genetics , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli Proteins/genetics , Genetic Complementation Test , Molecular Dynamics Simulation , Peptide Elongation Factors/genetics , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Rho Factor/chemistry , Rho Factor/genetics , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Species Specificity , Structure-Activity Relationship , Thermotoga maritima/genetics , Transcription Factors/genetics
16.
PLoS One ; 5(4): e10013, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20368803

ABSTRACT

BACKGROUND: The removal of introns from pre-mRNA is carried out by a large macromolecular machine called the spliceosome. The peptidyl-prolyl cis/trans isomerase PPIL1 is a component of the human spliceosome and binds to the spliceosomal SKIP protein via a binding site distinct from its active site. PRINCIPAL FINDINGS: Here, we have studied the PPIL1 protein and its interaction with SKIP biochemically and by X-ray crystallography. A minimal linear binding epitope derived from the SKIP protein could be determined using a peptide array. A 36-residue region of SKIP centred on an eight-residue epitope suffices to bind PPIL1 in pull-down experiments. The crystal structure of PPIL1 in complex with the inhibitor cyclosporine A (CsA) was obtained at a resolution of 1.15 A and exhibited two bound Cd(2+) ions that enabled SAD phasing. PPIL1 residues that have previously been implicated in binding of SKIP are involved in the coordination of Cd(2+) ions in the present crystal structure. Employing the present crystal structure, the determined minimal binding epitope and previously published NMR data, a molecular docking study was performed. In the docked model of the PPIL1.SKIP interaction, a proline residue of SKIP is buried in a hydrophobic pocket of PPIL1. This hydrophobic contact is encircled by several hydrogen bonds between the SKIP peptide and PPIL1. CONCLUSION: We characterized a short, linear epitope of SKIP that is sufficient to bind the PPIL1 protein. Our data indicate that this SKIP peptide could function in recruiting PPIL1 into the core of the spliceosome. We present a molecular model for the binding mode of SKIP to PPIL1 which emphasizes the versatility of cyclophilin-type PPIases to engage in additional interactions with other proteins apart from active site contacts despite their limited surface area.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Cyclosporine/chemistry , Peptidylprolyl Isomerase/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Binding Sites , Cadmium/chemistry , Crystallography, X-Ray , Cyclosporine/metabolism , Epitopes , Humans , Immunosuppressive Agents , Peptidylprolyl Isomerase/metabolism , Protein Binding , Protein Conformation
17.
Biochemistry ; 46(37): 10685-93, 2007 Sep 18.
Article in English | MEDLINE | ID: mdl-17718519

ABSTRACT

Clostridial neurotoxins are the causative agents of the neuroparalytic disease botulism and tetanus. They block neurotransmitter release through specific proteolysis of one of the three soluble N-ethylmaleimide-sensitive-factor attachment protein receptors (SNAREs) SNAP-25, syntaxin, and synaptobrevin, which constitute part of the synaptic vesicle fusion machinery. The catalytic component of the clostridial neurotoxins is their light chain (LC), a Zn2+ endopeptidase. There are seven structurally and functionally related botulinum neurotoxins (BoNTs), termed serotype A to G, and tetanus neurotoxin (TeNT). Each of them exhibits unique specificity for their target SNAREs and peptide bond(s) they cleave. The mechanisms of action for substrate recognition and target cleavage are largely unknown. Here, we report structural and biochemical studies of BoNT/C1-LC, which is unique among BoNTs in that it exhibits dual specificity toward both syntaxin and SNAP-25. A distinct pocket (S1') near the active site likely achieves the correct register for the cleavage site by only allowing Ala as the P1' residue for both SNAP-25 and syntaxin. Mutations of this SNAP-25 residue dramatically reduce enzymatic activity. The remote alpha-exosite that was previously identified in the complex of BoNT/A-LC and SNAP-25 is structurally conserved in BoNT/C1. However, mutagenesis experiments show that the alpha-exosite of BoNT/C1 plays a less stringent role in substrate discrimination in comparison to that of BoNT/A, which could account for its dual substrate specificity.


Subject(s)
Botulinum Toxins/chemistry , Peptide Hydrolases/chemistry , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Qa-SNARE Proteins/chemistry , Rats , Sequence Alignment , Structure-Activity Relationship , Substrate Specificity , Synaptosomal-Associated Protein 25/chemistry
18.
J Biol Chem ; 282(7): 5004-5014, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17092934

ABSTRACT

An efficient research strategy integrating empirically guided, structure-based modeling and chemoinformatics was used to discover potent small molecule inhibitors of the botulinum neurotoxin serotype A light chain. First, a modeled binding mode for inhibitor 2-mercapto-3-phenylpropionyl-RATKML (K(i) = 330 nM) was generated, and required the use of a molecular dynamic conformer of the enzyme displaying the reorientation of surface loops bordering the substrate binding cleft. These flexible loops are conformationally variable in x-ray crystal structures, and the model predicted that they were pivotal for providing complementary binding surfaces and solvent shielding for the pseudo-peptide. The docked conformation of 2-mercapto-3-phenylpropionyl-RATKML was then used to refine our pharmacophore for botulinum serotype A light chain inhibition. Data base search queries derived from the pharmacophore were employed to mine small molecule (non-peptidic) inhibitors from the National Cancer Institute's Open Repository. Four of the inhibitors possess K(i) values ranging from 3.0 to 10.0 microM. Of these, NSC 240898 is a promising lead for therapeutic development, as it readily enters neurons, exhibits no neuronal toxicity, and elicits dose-dependent protection of synaptosomal-associated protein (of 25 kDa) in a primary culture of embryonic chicken neurons. Isothermal titration calorimetry showed that the interaction between NSC 240898 and the botulinum A light chain is largely entropy-driven, and occurs with a 1:1 stoichiometry and a dissociation constant of 4.6 microM.


Subject(s)
Botulinum Toxins, Type A/chemistry , Metalloproteases/chemistry , Models, Molecular , Neurons/chemistry , Protease Inhibitors/chemistry , Animals , Botulinum Toxins, Type A/metabolism , Botulism/drug therapy , Botulism/enzymology , Cells, Cultured , Chick Embryo , Metalloproteases/metabolism , Neurons/enzymology , Protease Inhibitors/metabolism , Protease Inhibitors/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL