Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Mol Biol ; 435(18): 168215, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37516426

ABSTRACT

Immunoglobulin light chain amyloidosis (AL) is caused by the aberrant production of amyloidogenic light chains (LC) that accumulate as amyloid deposits in vital organs. Distinct LC sequences in each patient yield distinct amyloid structures. However different tissue microenvironments may also cause identical protein precursors to adopt distinct amyloid structures. To address the impact of the tissue environment on the structural polymorphism of amyloids, we extracted fibrils from the kidney of an AL patient (AL55) whose cardiac amyloid structure was previously determined by our group. Here we show that the 4.0 Å resolution cryo-EM structure of the renal fibril is virtually identical to that reported for the cardiac fibril. These results provide the first structural evidence that LC amyloids independently deposited in different organs of the same AL patient share a common fold.


Subject(s)
Amyloid , Immunoglobulin Light-chain Amyloidosis , Humans , Amyloid/chemistry , Cryoelectron Microscopy/methods , Immunoglobulin Light-chain Amyloidosis/metabolism , Kidney/metabolism , Tumor Microenvironment
2.
Nucleic Acids Res ; 51(15): 8237-8254, 2023 08 25.
Article in English | MEDLINE | ID: mdl-37378428

ABSTRACT

Specificity in protein-DNA recognition arises from the synergy of several factors that stem from the structural and chemical signatures encoded within the targeted DNA molecule. Here, we deciphered the nature of the interactions driving DNA recognition and binding by the bacterial transcription factor PdxR, a member of the MocR family responsible for the regulation of pyridoxal 5'-phosphate (PLP) biosynthesis. Single particle cryo-EM performed on the PLP-PdxR bound to its target DNA enabled the isolation of three conformers of the complex, which may be considered as snapshots of the binding process. Moreover, the resolution of an apo-PdxR crystallographic structure provided a detailed description of the transition of the effector domain to the holo-PdxR form triggered by the binding of the PLP effector molecule. Binding analyses of mutated DNA sequences using both wild type and PdxR variants revealed a central role of electrostatic interactions and of the intrinsic asymmetric bending of the DNA in allosterically guiding the holo-PdxR-DNA recognition process, from the first encounter through the fully bound state. Our results detail the structure and dynamics of the PdxR-DNA complex, clarifying the mechanism governing the DNA-binding mode of the holo-PdxR and the regulation features of the MocR family of transcription factors.


Subject(s)
Bacterial Proteins , Transcription Factors , Bacteria/genetics , Bacterial Proteins/metabolism , DNA/metabolism , Protein Binding , Pyridoxal Phosphate/metabolism , Transcription Factors/metabolism , Bacillus clausii/genetics
3.
Nat Commun ; 14(1): 239, 2023 01 16.
Article in English | MEDLINE | ID: mdl-36646699

ABSTRACT

hnRNPDL is a ribonucleoprotein (RNP) involved in transcription and RNA-processing that hosts missense mutations causing limb-girdle muscular dystrophy D3 (LGMD D3). Mammalian-specific alternative splicing (AS) renders three natural isoforms, hnRNPDL-2 being predominant in humans. We present the cryo-electron microscopy structure of full-length hnRNPDL-2 amyloid fibrils, which are stable, non-toxic, and bind nucleic acids. The high-resolution amyloid core consists of a single Gly/Tyr-rich and highly hydrophilic filament containing internal water channels. The RNA binding domains are located as a solenoidal coat around the core. The architecture and activity of hnRNPDL-2 fibrils are reminiscent of functional amyloids, our results suggesting that LGMD D3 might be a loss-of-function disease associated with impaired fibrillation. Strikingly, the fibril core matches exon 6, absent in the soluble hnRNPDL-3 isoform. This provides structural evidence for AS controlling hnRNPDL assembly by precisely including/skipping an amyloid exon, a mechanism that holds the potential to generate functional diversity in RNPs.


Subject(s)
Amyloid , Muscular Dystrophies, Limb-Girdle , Ribonucleoproteins , Humans , Alternative Splicing , Amyloid/metabolism , Amyloidogenic Proteins/metabolism , Cryoelectron Microscopy , Muscular Dystrophies, Limb-Girdle/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Ribonucleoproteins/metabolism
4.
Mol Ther ; 31(2): 362-373, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36114671

ABSTRACT

The uneven worldwide vaccination coverage against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and emergence of variants escaping immunity call for broadly effective and easily deployable therapeutic agents. We have previously described the human single-chain scFv76 antibody, which recognizes SARS-CoV-2 Alpha, Beta, Gamma and Delta variants. We now show that scFv76 also neutralizes the infectivity and fusogenic activity of the Omicron BA.1 and BA.2 variants. Cryoelectron microscopy (cryo-EM) analysis reveals that scFv76 binds to a well-conserved SARS-CoV-2 spike epitope, providing the structural basis for its broad-spectrum activity. We demonstrate that nebulized scFv76 has therapeutic efficacy in a severe hACE2 transgenic mouse model of coronavirus disease 2019 (COVID-19) pneumonia, as shown by body weight and pulmonary viral load data. Counteraction of infection correlates with inhibition of lung inflammation, as observed by histopathology and expression of inflammatory cytokines and chemokines. Biomarkers of pulmonary endothelial damage were also significantly reduced in scFv76-treated mice. The results support use of nebulized scFv76 for COVID-19 induced by any SARS-CoV-2 variants that have emerged so far.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Animals , Mice , SARS-CoV-2/genetics , Cryoelectron Microscopy , Respiratory Aerosols and Droplets , Antibodies , Mice, Transgenic , Lung , Antibodies, Viral , Antibodies, Neutralizing
5.
Nat Commun ; 13(1): 7041, 2022 11 17.
Article in English | MEDLINE | ID: mdl-36396658

ABSTRACT

AA amyloidosis is a systemic disease characterized by deposition of misfolded serum amyloid A protein (SAA) into cross-ß amyloid in multiple organs in humans and animals. AA amyloidosis occurs at high SAA serum levels during chronic inflammation. Prion-like transmission was reported as possible cause of extreme AA amyloidosis prevalence in captive animals, e.g. 70% in cheetah and 57-73% in domestic short hair (DSH) cats kept in zoos and shelters, respectively. Herein, we present the 3.3 Å cryo-EM structure of AA amyloid extracted post-mortem from the kidney of a DSH cat with renal failure, deceased in a shelter with extreme disease prevalence. The structure reveals a cross-ß architecture assembled from two 76-residue long proto-filaments. Despite >70% sequence homology to mouse and human SAA, the cat SAA variant adopts a distinct amyloid fold. Inclusion of an eight-residue insert unique to feline SAA contributes to increased amyloid stability. The presented feline AA amyloid structure is fully compatible with the 99% identical amino acid sequence of amyloid fragments of captive cheetah.


Subject(s)
Acinonyx , Amyloidosis , Animals , Cats , Mice , Acinonyx/metabolism , Amyloid/metabolism , Amyloidosis/metabolism , Cryoelectron Microscopy , Prevalence , Serum Amyloid A Protein/metabolism
6.
Viruses ; 14(6)2022 05 27.
Article in English | MEDLINE | ID: mdl-35746629

ABSTRACT

Pyridobenzothiazolone derivatives are a promising class of broad-spectrum antivirals. However, the mode of action of these compounds remains poorly understood. The HeE1-17Y derivative has already been shown to be a potent compound against a variety of flaviviruses of global relevance. In this work, the mode of action of HeE1-17Y has been studied for West Nile virus taking advantage of reporter replication particles (RRPs). Viral infectivity was drastically reduced by incubating the compound with the virus before infection, thus suggesting a direct interaction with the viral particles. Indeed, RRPs incubated with the inhibitor appeared to be severely compromised in electron microscopy analysis. HeE1-17Y is active against other enveloped viruses, including SARS-CoV-2, but not against two non-enveloped viruses, suggesting a virucidal mechanism that involves the alteration of the viral membrane.


Subject(s)
COVID-19 , Flavivirus , RNA Viruses , Viruses , Antiviral Agents/pharmacology , Humans , SARS-CoV-2
7.
Int J Biol Macromol ; 193(Pt A): 401-413, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34673109

ABSTRACT

The trimeric CCAAT-binding NF-Y is a "pioneer" Transcription Factor -TF- known to cooperate with neighboring TFs to regulate gene expression. Genome-wide analyses detected a precise stereo-alignment -10/12 bp- of CCAAT with E-box elements and corresponding colocalization of NF-Y with basic-Helix-Loop-Helix (bHLH) TFs. We dissected here NF-Y interactions with USF1 and MAX. USF1, but not MAX, cooperates in DNA binding with NF-Y. NF-Y and USF1 synergize to activate target promoters. Reconstruction of complexes by structural means shows independent DNA binding of MAX, whereas USF1 has extended contacts with NF-Y, involving the USR, a USF-specific amino acid sequence stretch required for trans-activation. The USR is an intrinsically disordered domain and adopts different conformations based on E-box-CCAAT distances. Deletion of the USR abolishes cooperative DNA binding with NF-Y. Our data indicate that the functionality of certain unstructured domains involves adapting to small variation in stereo-alignments of the multimeric TFs sites.


Subject(s)
DNA/metabolism , Upstream Stimulatory Factors/metabolism , Gene Expression Regulation , Humans , Promoter Regions, Genetic , Protein Binding , Protein Domains
8.
Mol Cell ; 81(14): 2929-2943.e6, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34166608

ABSTRACT

The HCN1-4 channel family is responsible for the hyperpolarization-activated cation current If/Ih that controls automaticity in cardiac and neuronal pacemaker cells. We present cryoelectron microscopy (cryo-EM) structures of HCN4 in the presence or absence of bound cAMP, displaying the pore domain in closed and open conformations. Analysis of cAMP-bound and -unbound structures sheds light on how ligand-induced transitions in the channel cytosolic portion mediate the effect of cAMP on channel gating and highlights the regulatory role of a Mg2+ coordination site formed between the C-linker and the S4-S5 linker. Comparison of open/closed pore states shows that the cytosolic gate opens through concerted movements of the S5 and S6 transmembrane helices. Furthermore, in combination with molecular dynamics analyses, the open pore structures provide insights into the mechanisms of K+/Na+ permeation. Our results contribute mechanistic understanding on HCN channel gating, cyclic nucleotide-dependent modulation, and ion permeation.


Subject(s)
Cell Membrane Permeability/physiology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Ion Channel Gating/physiology , Ions/metabolism , Muscle Proteins/metabolism , Potassium Channels/metabolism , Cell Line , Cryoelectron Microscopy/methods , Cyclic AMP/metabolism , HEK293 Cells , Humans
9.
J Mol Biol ; 433(18): 167119, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34181981

ABSTRACT

The E2F1 transcription factor is a master regulator of cell-cycle progression whose uncontrolled activation contributes to tumor cells growth. E2F1 binds DNA as a heterodimer with DP partners, resulting in a multi-domain quaternary-structure complex composed of DNA binding domains, a coiled coil domain and a marked box domain separated by short linkers. Building on the 3D knowledge of the single domains of E2F and DPs, we characterized the structure and dynamics of the complete E2F1/DP1/DNA complex by a combination of small-angle X-ray scattering and molecular dynamics simulations. It shows an asymmetric contribution of the dynamics of the two proteins. Namely, the coiled-coil domain leans toward the DP1 side of the complex; the DP1 loop between α2 and α3 of the DBD partially populates a helical structure leaning far from the DNA and in the same direction of the coiled-coil domain; and the N-terminal disordered region of DP1, rich in basic residues, contributes to DNA binding stabilization. Intriguingly, tumor mutations in the flexible regions of the complex suggest that perturbation of protein dynamics could affect protein function in a context-dependent way. Our data suggest fundamental contributions of DP proteins in distinct aspects of E2F biology.


Subject(s)
DNA/chemistry , DNA/metabolism , E2F1 Transcription Factor/chemistry , E2F1 Transcription Factor/metabolism , Transcription Factor DP1/chemistry , Transcription Factor DP1/metabolism , Cell Cycle , Humans , Models, Molecular , Molecular Dynamics Simulation , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Nucleic Acid Conformation , Phosphorylation , Protein Binding , Protein Conformation
10.
Eur Heart J ; 42(28): 2780-2792, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34104945

ABSTRACT

AIMS: Increased shedding of extracellular vesicles (EVs)-small, lipid bilayer-delimited particles with a role in paracrine signalling-has been associated with human pathologies, e.g. atherosclerosis, but whether this is true for cardiac diseases is unknown. METHODS AND RESULTS: Here, we used the surface antigen CD172a as a specific marker of cardiomyocyte (CM)-derived EVs; the CM origin of CD172a+ EVs was supported by their content of cardiac-specific proteins and heart-enriched microRNAs. We found that patients with aortic stenosis, ischaemic heart disease, or cardiomyopathy had higher circulating CD172a+ cardiac EV counts than did healthy subjects. Cellular stress was a major determinant of EV release from CMs, with hypoxia increasing shedding in in vitro and in vivo experiments. At the functional level, EVs isolated from the supernatant of CMs derived from human-induced pluripotent stem cells and cultured in a hypoxic atmosphere elicited a positive inotropic response in unstressed CMs, an effect we found to be dependent on an increase in the number of EVs expressing ceramide on their surface. Of potential clinical relevance, aortic stenosis patients with the highest counts of circulating cardiac CD172a+ EVs had a more favourable prognosis for transcatheter aortic valve replacement than those with lower counts. CONCLUSION: We identified circulating CD172a+ EVs as cardiac derived, showing their release and function and providing evidence for their prognostic potential in aortic stenosis patients.


Subject(s)
Extracellular Vesicles , MicroRNAs , Myocardial Infarction , Humans , Hypoxia , Myocardium , Myocytes, Cardiac
11.
Plant J ; 105(1): 49-61, 2021 01.
Article in English | MEDLINE | ID: mdl-33098724

ABSTRACT

NF-Y transcription factor comprises three subunits: NF-YA, NF-YB and NF-YC. NF-YB and NF-YC dimerize through their histone fold domain (HFD), which can bind DNA in a non-sequence-specific fashion while serving as a scaffold for NF-YA trimerization. Upon trimerization, NF-YA specifically recognizes the CCAAT box sequence on promoters and enhancers. In plants, each NF-Y subunit is encoded by several genes giving rise to hundreds of potential heterotrimeric combinations. In addition, plant NF-YBs and NF-YCs interact with other protein partners to recognize a plethora of genomic motifs, as the CCT protein family that binds CORE sites. The NF-Y subunit organization and its DNA-binding properties, together with the NF-Y HFD capacity to adapt different protein modules, represent plant-specific features that play a key role in development, growth and reproduction. Despite their relevance, these features are still poorly understood at the molecular level. Here, we present the structures of Arabidopsis and rice NF-YB/NF-YC dimers, and of an Arabidopsis NF-Y trimer in complex with the FT CCAAT box, together with biochemical data on NF-Y mutants. The dimeric structures identify the key residues for NF-Y HFD stabilization. The NF-Y/DNA structure and the mutation experiments shed light on HFD trimerization interface properties and the NF-YA sequence appetite for the bases flanking the CCAAT motif. These data explain the logic of plant NF-Y gene expansion: the trimerization adaptability and the flexible DNA-binding rules serve the scopes of accommodating the large number of NF-YAs, CCTs and possibly other NF-Y HFD binding partners and a diverse audience of genomic motifs.


Subject(s)
Arabidopsis Proteins/metabolism , CCAAT-Binding Factor/metabolism , DNA, Plant/metabolism , Amino Acid Sequence , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Binding Sites , CCAAT-Binding Factor/chemistry , CCAAT-Binding Factor/genetics , DNA, Plant/chemistry , Dimerization , Oryza/genetics , Oryza/metabolism , Plant Proteins/chemistry , Plant Proteins/genetics , Plant Proteins/metabolism , Protein Structure, Tertiary
12.
Cells ; 9(11)2020 10 29.
Article in English | MEDLINE | ID: mdl-33138093

ABSTRACT

NF-Y is a transcription factor (TF) comprising three subunits (NF-YA, NF-YB, NF-YC) that binds with high specificity to the CCAAT sequence, a widespread regulatory element in gene promoters of prosurvival, cell-cycle-promoting, and metabolic genes. Tumor cells undergo "metabolic rewiring" through overexpression of genes involved in such pathways, many of which are under NF-Y control. In addition, NF-YA appears to be overexpressed in many tumor types. Thus, limiting NF-Y activity may represent a desirable anti-cancer strategy, which is an ongoing field of research. With virtual-screening docking simulations on a library of pharmacologically active compounds, we identified suramin as a potential NF-Y inhibitor. We focused on suramin given its high water-solubility that is an important factor for in vitro testing, since NF-Y is sensitive to DMSO. By electrophoretic mobility shift assays (EMSA), isothermal titration calorimetry (ITC), STD NMR, X-ray crystallography, and molecular dynamics (MD) simulations, we showed that suramin binds to the histone fold domains (HFDs) of NF-Y, preventing DNA-binding. Our analyses, provide atomic-level detail on the interaction between suramin and NF-Y and reveal a region of the protein, nearby the suramin-binding site and poorly conserved in other HFD-containing TFs, that may represent a promising starting point for rational design of more specific and potent inhibitors with potential therapeutic applications.


Subject(s)
CCAAT-Binding Factor/antagonists & inhibitors , CCAAT-Binding Factor/chemistry , Suramin/chemistry , Suramin/pharmacology , Transcription Factors/antagonists & inhibitors , Transcription Factors/chemistry , Amino Acid Sequence , Biophysical Phenomena , DNA/metabolism , Humans , Magnetic Resonance Spectroscopy , Molecular Dynamics Simulation , Protein Multimerization , Structure-Activity Relationship
13.
Acta Crystallogr F Struct Biol Commun ; 76(Pt 9): 392-397, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32880586

ABSTRACT

The unintentional crystallization of contaminant proteins in the place of target recombinant proteins is sporadically reported, despite the availability of stringent expression/purification protocols and of software for the detection of contaminants. Typically, the contaminant protein originates from the expression organism (for example Escherichia coli), but in rare circumstances contaminants from different sources have been reported. Here, a case of contamination from a Serratia bacterial strain that occurred while attempting to crystallize an unrelated protein from Burkholderia pseudomallei (overexpressed in E. coli) is presented. The contamination led to the unintended crystallization and structure analysis of a cyanase hydratase from a bacterial strain of the Serratia genus, an opportunistic enterobacterium that grows under conditions similar to those of E. coli and that is found in a variety of habitats, including the laboratory environment. In this context, the procedures that were adopted to identify the contaminant based on crystallographic data only are presented and the crystal structure of Serrata spp. cyanase hydratase is briefly discussed.


Subject(s)
Artifacts , Crystallography, X-Ray/standards , Cyanates/chemistry , Escherichia coli/genetics , Hydro-Lyases/ultrastructure , Binding Sites , Burkholderia pseudomallei/enzymology , Burkholderia pseudomallei/genetics , Cyanates/metabolism , Escherichia coli/enzymology , Gene Expression , Humans , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Serratia/enzymology , Serratia/genetics , Transgenes
14.
J Biol Chem ; 295(49): 16572-16584, 2020 12 04.
Article in English | MEDLINE | ID: mdl-32952127

ABSTRACT

Amyloid fibrils are polymeric structures originating from aggregation of misfolded proteins. In vivo, proteolysis may modulate amyloidogenesis and fibril stability. In light chain (AL) amyloidosis, fragmented light chains (LCs) are abundant components of amyloid deposits; however, site and timing of proteolysis are debated. Identification of the N and C termini of LC fragments is instrumental to understanding involved processes and enzymes. We investigated the N and C terminome of the LC proteoforms in fibrils extracted from the hearts of two AL cardiomyopathy patients, using a proteomic approach based on derivatization of N- and C-terminal residues, followed by mapping of fragmentation sites on the structures of native and fibrillar relevant LCs. We provide the first high-specificity map of proteolytic cleavages in natural AL amyloid. Proteolysis occurs both on the LC variable and constant domains, generating a complex fragmentation pattern. The structural analysis indicates extensive remodeling by multiple proteases, largely taking place on poorly folded regions of the fibril surfaces. This study adds novel important knowledge on amyloid LC processing: although our data do not exclude that proteolysis of native LC dimers may destabilize their structure and favor fibril formation, the data show that LC deposition largely precedes the proteolytic events documentable in mature AL fibrils.


Subject(s)
Amyloid/chemistry , Immunoglobulin Light-chain Amyloidosis/pathology , Myocardium/metabolism , Amino Acid Sequence , Amyloid/metabolism , Chromatography, High Pressure Liquid , Electrophoresis, Gel, Two-Dimensional , Humans , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/metabolism , Immunoglobulin Light-chain Amyloidosis/metabolism , Peptides/analysis , Protein Structure, Secondary , Protein Structure, Tertiary , Proteolysis , Tandem Mass Spectrometry
15.
Plant Physiol ; 182(4): 2143-2153, 2020 04.
Article in English | MEDLINE | ID: mdl-32015077

ABSTRACT

Plant growth largely depends on the maintenance of adequate intracellular levels of potassium (K+). The families of 10 Calcineurin B-Like (CBL) calcium sensors and 26 CBL-Interacting Protein Kinases (CIPKs) of Arabidopsis (Arabidopsis thaliana) decode the calcium signals elicited by environmental inputs to regulate different ion channels and transporters involved in the control of K+ fluxes by phosphorylation-dependent and -independent events. However, the detailed molecular mechanisms governing target specificity require investigation. Here, we show that the physical interaction between CIPK23 and the noncanonical ankyrin domain in the cytosolic side of the inward-rectifier K+ channel AKT1 regulates kinase docking and channel activation. Point mutations on this domain specifically alter binding to CIPK23, enhancing or impairing the ability of CIPK23 to regulate channel activity. Our data demonstrate the relevance of this protein-protein interaction that contributes to the formation of a complex between CIPK23/CBL1 and AKT1 in the membrane for the proper regulation of K+ transport.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Calcium-Binding Proteins/metabolism , Potassium Channels/metabolism , Protein Serine-Threonine Kinases/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Calcium-Binding Proteins/genetics , Point Mutation , Potassium/metabolism , Potassium Channels/genetics , Protein Serine-Threonine Kinases/genetics
16.
FASEB J ; 34(1): 1182-1197, 2020 01.
Article in English | MEDLINE | ID: mdl-31914658

ABSTRACT

d-Amino acids are the "wrong" enantiomers of amino acids as they are not used in proteins synthesis but evolved in selected functions. On this side, d-aspartate (d-Asp) plays several significant roles in mammals, especially as an agonist of N-methyl-d-aspartate receptors (NMDAR), and is involved in relevant diseases, such as schizophrenia and Alzheimer's disease. In vivo modulation of d-Asp levels represents an intriguing task to cope with such pathological states. As little is known about d-Asp synthesis, the only option for modulating the levels is via degradation, which is due to the flavoenzyme d-aspartate oxidase (DASPO). Here we present the first three-dimensional structure of a DASPO enzyme (from human) which belongs to the d-amino acid oxidase family. Notably, human DASPO differs from human d-amino acid oxidase (attributed to d-serine degradation, the main coagonist of NMDAR) showing peculiar structural features (a specific active site charge distribution), oligomeric state and kinetic mechanism, and a higher FAD affinity and activity. These results provide useful insights into the structure-function relationships of human DASPO: modulating its activity represents now a feasible novel therapeutic target.


Subject(s)
Brain/metabolism , D-Aspartate Oxidase/chemistry , D-Aspartate Oxidase/metabolism , D-Aspartic Acid/analysis , Animals , Antipsychotic Agents/pharmacology , Binding Sites , Cattle , Crystallography, X-Ray , Dimerization , Drug Design , Humans , Kinetics , Ligands , Mice , Molecular Docking Simulation , Mutagenesis, Site-Directed , Protein Binding , Receptors, N-Methyl-D-Aspartate/metabolism , Substrate Specificity , Swine
17.
Mol Cell ; 76(6): 922-937.e7, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31604602

ABSTRACT

In the arms race against bacteria, bacteriophages have evolved diverse anti-CRISPR proteins (Acrs) that block CRISPR-Cas immunity. Acrs play key roles in the molecular coevolution of bacteria with their predators, use a variety of mechanisms of action, and provide tools to regulate Cas-based genome manipulation. Here, we present structural and functional analyses of AcrIIA6, an Acr from virulent phages, exploring its unique anti-CRISPR action. Our cryo-EM structures and functional data of AcrIIA6 binding to Streptococcus thermophilus Cas9 (St1Cas9) show that AcrIIA6 acts as an allosteric inhibitor and induces St1Cas9 dimerization. AcrIIA6 reduces St1Cas9 binding affinity for DNA and prevents DNA binding within cells. The PAM and AcrIIA6 recognition sites are structurally close and allosterically linked. Mechanistically, AcrIIA6 affects the St1Cas9 conformational dynamics associated with PAM binding. Finally, we identify a natural St1Cas9 variant resistant to AcrIIA6 illustrating Acr-driven mutational escape and molecular diversification of Cas9 proteins.


Subject(s)
Bacteriophages/metabolism , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , DNA/metabolism , Streptococcus thermophilus/enzymology , Viral Proteins/metabolism , Allosteric Regulation , Bacteriophages/genetics , Binding Sites , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/ultrastructure , DNA/genetics , DNA/ultrastructure , Escherichia coli/enzymology , Escherichia coli/genetics , Humans , K562 Cells , Kinetics , Mutation , Protein Binding , Protein Conformation , Streptococcus thermophilus/genetics , Structure-Activity Relationship , Viral Proteins/genetics , Viral Proteins/ultrastructure
18.
J Mol Biol ; 431(22): 4523-4526, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31473159

ABSTRACT

Bacterial NADPH-dependent glutamate synthase (GltS) is a complex iron-sulfur flavoprotein that catalyzes the reductive synthesis of two L-Glu molecules from L-Gln and 2-oxo-glutarate. GltS functional unit hosts an α-subunit (αGltS) and a ß-subunit (ßGltS) that assemble in different αß oligomers in solution. Here, we present the cryo-electron microscopy structures of Azospirillum brasilense GltS in four different oligomeric states (α4ß3, α4ß4, α6ß4 and α6ß6, in the 3.5- to 4.1-Å resolution range). Our study provides a comprehensive GltS model that details the inter-protomeric assemblies and allows unequivocal location of the FAD cofactor and of two electron transfer [4Fe-4S]+1,+2 clusters within ßGltS.


Subject(s)
Azospirillum brasilense/enzymology , Cryoelectron Microscopy/methods , Glutamate Synthase/metabolism , Glutamate Synthase/ultrastructure , Catalysis , Electron Transport , Flavin Mononucleotide/metabolism , Flavin-Adenine Dinucleotide/metabolism , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/ultrastructure
19.
J Med Chem ; 61(14): 5910-5921, 2018 Jul 26.
Article in English | MEDLINE | ID: mdl-29966094

ABSTRACT

Protein-protein interactions (PPIs) are known to play an essential role between the neuronal calcium sensor 1 (NCS-1) and the guanine exchange factor Ric8a to regulate synapse function, emerging as a druggable interface for synaptopathies such as the fragile X syndrome (FXS). Recently, the phenothiazine FD44 has been identified as an inhibitor of this PPI, decreasing the abnormally high synapse number and enhancing associative learning in a FXS animal model. Here, we have integrated advanced experimental and computational studies to obtain important structural insights into Drosophila NCS-1/FD44 recognition to understand the basis of its affinity and specificity and generate improved PPI regulators. This has allowed the identification of a new small drug-like molecule, IGS-1.76, which efficiently inhibits the human NCS-1/Ric8a complex with improved binding potency. The crystal structure of the Drosophila NCS-1/IGS-1.76 complex demonstrates that the new inhibitor, although chemically different from FD44, shares the same mechanism of action and constitutes a new hit candidate for FXS.


Subject(s)
Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Neuronal Calcium-Sensor Proteins/antagonists & inhibitors , Neuropeptides/antagonists & inhibitors , Phenothiazines/pharmacology , Synapses/drug effects , Synapses/physiology , Guanine Nucleotide Exchange Factors/metabolism , Humans , Molecular Dynamics Simulation , Neuronal Calcium-Sensor Proteins/chemistry , Neuronal Calcium-Sensor Proteins/metabolism , Neuropeptides/chemistry , Neuropeptides/metabolism , Protein Conformation, alpha-Helical
20.
Plant Cell ; 29(10): 2570-2580, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28970335

ABSTRACT

Plants acquire potassium (K+) ions for cell growth and movement via regulated diffusion through K+ channels. Here, we present crystallographic and functional data showing that the K+ inward rectifier KAT1 (K+Arabidopsis thaliana 1) channel is regulated by 14-3-3 proteins and further modulated by the phytotoxin fusicoccin, in analogy to the H+-ATPase. We identified a 14-3-3 mode III binding site at the very C terminus of KAT1 and cocrystallized it with tobacco (Nicotiana tabacum) 14-3-3 proteins to describe the protein complex at atomic detail. Validation of this interaction by electrophysiology shows that 14-3-3 binding augments KAT1 conductance by increasing the maximal current and by positively shifting the voltage dependency of gating. Fusicoccin potentiates the 14-3-3 effect on KAT1 activity by stabilizing their interaction. Crystal structure of the ternary complex reveals a noncanonical binding site for the toxin that adopts a novel conformation. The structural insights underscore the adaptability of fusicoccin, predicting more potential targets than so far anticipated. The data further advocate a common mechanism of regulation of the proton pump and a potassium channel, two essential elements in K+ uptake in plant cells.


Subject(s)
14-3-3 Proteins/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Glycosides/pharmacology , Arabidopsis/drug effects , Electrophysiology , Plant Proteins/metabolism , Potassium Channels, Inwardly Rectifying/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...