Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
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
2.
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
3.
Mol Cell ; 65(2): 247-259, 2017 Jan 19.
Article in English | MEDLINE | ID: mdl-27986371

ABSTRACT

Monoubiquitination and deubiquitination of FANCD2:FANCI heterodimer is central to DNA repair in a pathway that is defective in the cancer predisposition syndrome Fanconi anemia (FA). The "FA core complex" contains the RING-E3 ligase FANCL and seven other essential proteins that are mutated in various FA subtypes. Here, we purified recombinant FA core complex to reveal the function of these other proteins. The complex contains two spatially separate FANCL molecules that are dimerized by FANCB and FAAP100. FANCC and FANCE act as substrate receptors and restrict monoubiquitination to the FANCD2:FANCI heterodimer in only a DNA-bound form. FANCA and FANCG are dispensable for maximal in vitro ubiquitination. Finally, we show that the reversal of this reaction by the USP1:UAF1 deubiquitinase only occurs when DNA is disengaged. Our work reveals the mechanistic basis for temporal and spatial control of FANCD2:FANCI monoubiquitination that is critical for chemotherapy responses and prevention of Fanconi anemia.


Subject(s)
Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Fanconi Anemia/metabolism , Ubiquitination , Cell Line , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/metabolism , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group A Protein/metabolism , Fanconi Anemia Complementation Group C Protein/metabolism , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group E Protein/metabolism , Fanconi Anemia Complementation Group G Protein/metabolism , Fanconi Anemia Complementation Group L Protein/metabolism , Fanconi Anemia Complementation Group Proteins/genetics , Humans , Inhibitor of Differentiation Protein 2/metabolism , Multiprotein Complexes , Nuclear Proteins/metabolism , Protein Binding , Protein Multimerization , Recombinant Proteins/metabolism , Substrate Specificity , Time Factors , Transfection , Ubiquitin-Specific Proteases/metabolism
4.
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
5.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Article in English | MEDLINE | ID: mdl-35115399

ABSTRACT

The RecQ-like helicase BLM cooperates with topoisomerase IIIα, RMI1, and RMI2 in a heterotetrameric complex (the "Bloom syndrome complex") for dissolution of double Holliday junctions, key intermediates in homologous recombination. Mutations in any component of the Bloom syndrome complex can cause genome instability and a highly cancer-prone disorder called Bloom syndrome. Some heterozygous carriers are also predisposed to breast cancer. To understand how the activities of BLM helicase and topoisomerase IIIα are coupled, we purified the active four-subunit complex. Chemical cross-linking and mass spectrometry revealed a unique architecture that links the helicase and topoisomerase domains. Using biochemical experiments, we demonstrated dimerization mediated by the N terminus of BLM with a 2:2:2:2 stoichiometry within the Bloom syndrome complex. We identified mutations that independently abrogate dimerization or association of BLM with RMI1, and we show that both are dysfunctional for dissolution using in vitro assays and cause genome instability and synthetic lethal interactions with GEN1/MUS81 in cells. Truncated BLM can also inhibit the activity of full-length BLM in mixed dimers, suggesting a putative mechanism of dominant-negative action in carriers of BLM truncation alleles. Our results identify critical molecular determinants of Bloom syndrome complex assembly required for double Holliday junction dissolution and maintenance of genome stability.


Subject(s)
Bloom Syndrome/genetics , DNA, Cruciform/genetics , Genomic Instability/genetics , Alleles , Carrier Proteins/genetics , Cell Line , DNA Topoisomerases, Type I/genetics , Humans , Mutation/genetics , Protein Binding/genetics , RecQ Helicases/genetics , Recombination, Genetic/genetics , Solubility
6.
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
7.
Int J Mol Sci ; 21(22)2020 Nov 16.
Article in English | MEDLINE | ID: mdl-33207833

ABSTRACT

In plant grana thylakoid membranes Photosystem II (PSII) associates with a variable number of antenna proteins (LHCII) to form different types of supercomplexes (PSII-LHCII), whose organization is dynamically adjusted in response to light cues, with the C2S2 more abundant in high-light and the C2S2M2 in low-light. Paired PSII-LHCII supercomplexes interacting at their stromal surface from adjacent thylakoid membranes were previously suggested to mediate grana stacking. Here, we present the cryo-electron microscopy maps of paired C2S2 and C2S2M2 supercomplexes isolated from pea plants grown in high-light and low-light, respectively. These maps show a different rotational offset between the two supercomplexes in the pair, responsible for modifying their reciprocal interaction and energetic connectivity. This evidence reveals a different way by which paired PSII-LHCII supercomplexes can mediate grana stacking at diverse irradiances. Electrostatic stromal interactions between LHCII trimers almost completely overlapping in the paired C2S2 can be the main determinant by which PSII-LHCII supercomplexes mediate grana stacking in plants grown in high-light, whereas the mutual interaction of stromal N-terminal loops of two facing Lhcb4 subunits in the paired C2S2M2 can fulfil this task in plants grown in low-light. The high-light induced accumulation of the Lhcb4.3 protein in PSII-LHCII supercomplexes has been previously reported. Our cryo-electron microscopy map at 3.8 Å resolution of the C2S2 supercomplex isolated from plants grown in high-light suggests the presence of the Lhcb4.3 protein revealing peculiar structural features of this high-light-specific antenna important for photoprotection.


Subject(s)
Light-Harvesting Protein Complexes/metabolism , Light , Photosystem II Protein Complex/metabolism , Pisum sativum/enzymology , Thylakoids/enzymology , Light-Harvesting Protein Complexes/chemistry , Photosystem II Protein Complex/chemistry
8.
J Virol ; 92(3)2018 02 01.
Article in English | MEDLINE | ID: mdl-29142132

ABSTRACT

Despite the availability of two attenuated vaccines, rotavirus (RV) gastroenteritis remains an important cause of mortality among children in developing countries, causing about 215,000 infant deaths annually. Currently, there are no specific antiviral therapies available. RV is a nonenveloped virus with a segmented double-stranded RNA genome. Viral genome replication and assembly of transcriptionally active double-layered particles (DLPs) take place in cytoplasmic viral structures called viroplasms. In this study, we describe strong impairment of the early stages of RV replication induced by a small molecule known as an RNA polymerase III inhibitor, ML-60218 (ML). This compound was found to disrupt already assembled viroplasms and to hamper the formation of new ones without the need for de novo transcription of cellular RNAs. This phenotype was correlated with a reduction in accumulated viral proteins and newly made viral genome segments, disappearance of the hyperphosphorylated isoforms of the viroplasm-resident protein NSP5, and inhibition of infectious progeny virus production. In in vitro transcription assays with purified DLPs, ML showed dose-dependent inhibitory activity, indicating the viral nature of its target. ML was found to interfere with the formation of higher-order structures of VP6, the protein forming the DLP outer layer, without compromising its ability to trimerize. Electron microscopy of ML-treated DLPs showed dose-dependent structural damage. Our data suggest that interactions between VP6 trimers are essential, not only for DLP stability, but also for the structural integrity of viroplasms in infected cells.IMPORTANCE Rotavirus gastroenteritis is responsible for a large number of infant deaths in developing countries. Unfortunately, in the countries where effective vaccines are urgently needed, the efficacy of the available vaccines is particularly low. Therefore, the development of antivirals is an important goal, as they might complement the available vaccines or represent an alternative option. Moreover, they may be decisive in fighting the acute phase of infection. This work describes the inhibitory effect on rotavirus replication of a small molecule initially reported as an RNA polymerase III inhibitor. The molecule is the first chemical compound identified that is able to disrupt viroplasms, the viral replication machinery, and to compromise the stability of DLPs by targeting the viral protein VP6. This molecule thus represents a starting point in the development of more potent and less cytotoxic compounds against rotavirus infection.


Subject(s)
RNA Polymerase III/antagonists & inhibitors , Rotavirus/physiology , Small Molecule Libraries/pharmacology , Viral Structures/drug effects , Animals , Cell Line , Chlorocebus aethiops , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Rotavirus/chemistry , Rotavirus/drug effects , Sf9 Cells , Viral Proteins/antagonists & inhibitors , Virus Replication/drug effects
9.
Proc Natl Acad Sci U S A ; 112(14): 4286-91, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25831490

ABSTRACT

RecQ helicases are a widely conserved family of ATP-dependent motors with diverse roles in nearly every aspect of bacterial and eukaryotic genome maintenance. However, the physical mechanisms by which RecQ helicases recognize and process specific DNA replication and repair intermediates are largely unknown. Here, we solved crystal structures of the human RECQ1 helicase in complexes with tailed-duplex DNA and ssDNA. The structures map the interactions of the ssDNA tail and the branch point along the helicase and Zn-binding domains, which, together with reported structures of other helicases, define the catalytic stages of helicase action. We also identify a strand-separating pin, which (uniquely in RECQ1) is buttressed by the protein dimer interface. A duplex DNA-binding surface on the C-terminal domain is shown to play a role in DNA unwinding, strand annealing, and Holliday junction (HJ) branch migration. We have combined EM and analytical ultracentrifugation approaches to show that RECQ1 can form what appears to be a flat, homotetrameric complex and propose that RECQ1 tetramers are involved in HJ recognition. This tetrameric arrangement suggests a platform for coordinated activity at the advancing and receding duplexes of an HJ during branch migration.


Subject(s)
DNA Helicases/chemistry , DNA/chemistry , RecQ Helicases/chemistry , Animals , Chromatography, Gel , Crystallization , Crystallography, X-Ray , DNA, Cruciform/physiology , DNA, Single-Stranded/chemistry , Escherichia coli/metabolism , Humans , Insecta , Molecular Conformation , Nucleic Acid Denaturation , Nucleotides/chemistry , Protein Binding , Protein Structure, Tertiary , Zinc/chemistry
10.
Bioorg Med Chem ; 20(1): 408-14, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22112537

ABSTRACT

Here, we report on the development of a novel methodology to aid in design of Hsp90 inhibitors, using molecular docking combined with artificial neural network (ANN) modelling. Inhibitors are first docked into the ATPase site of the Human Hsp90α crystal structures and the thermodynamic properties of the complexes together with various physical-chemical properties of the ligands are used as input to train a simple feed-forward, back propagation ANN, to predict the inhibitors' pIC(50)s. For an objective test set of 60 known Hsp90 inhibitors for which there are no crystallographic data available, the trained ANN is shown to give pIC(50)s accurate to within ±1 log unit, and the predictions are sufficiently good as to allow the majority of the inhibitors to be ranked correctly according to their potency.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Binding Sites , Computer Simulation , HSP90 Heat-Shock Proteins/metabolism , Humans , Ligands , Protein Structure, Tertiary , Quantitative Structure-Activity Relationship , Software , Thermodynamics
11.
Acta Crystallogr D Struct Biol ; 78(Pt 11): 1399-1411, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36322422

ABSTRACT

Oxygenic phototrophs perform carbon fixation through the Calvin-Benson cycle. Different mechanisms adjust the cycle and the light-harvesting reactions to rapid environmental changes. Photosynthetic glyceraldehyde 3-phosphate dehydrogenase (GAPDH) is a key enzyme in the cycle. In land plants, different photosynthetic GAPDHs exist: the most abundant isoform is formed by A2B2 heterotetramers and the least abundant by A4 homotetramers. Regardless of the subunit composition, GAPDH is the major consumer of photosynthetic NADPH and its activity is strictly regulated. While A4-GAPDH is regulated by CP12, AB-GAPDH is autonomously regulated through the C-terminal extension (CTE) of its B subunits. Reversible inhibition of AB-GAPDH occurs via the oxidation of a cysteine pair located in the CTE and the substitution of NADP(H) with NAD(H) in the cofactor-binding site. These combined conditions lead to a change in the oligomerization state and enzyme inhibition. SEC-SAXS and single-particle cryo-EM analysis were applied to reveal the structural basis of this regulatory mechanism. Both approaches revealed that spinach (A2B2)n-GAPDH oligomers with n = 1, 2, 4 and 5 co-exist in a dynamic system. B subunits mediate the contacts between adjacent tetramers in A4B4 and A8B8 oligomers. The CTE of each B subunit penetrates into the active site of a B subunit of the adjacent tetramer, which in turn moves its CTE in the opposite direction, effectively preventing the binding of the substrate 1,3-bisphosphoglycerate in the B subunits. The whole mechanism is made possible, and eventually controlled, by pyridine nucleotides. In fact, NAD(H), by removing NADP(H) from A subunits, allows the entrance of the CTE into the active site of the B subunit, hence stabilizing inhibited oligomers.


Subject(s)
NAD , Photosynthesis , NADP/chemistry , Scattering, Small Angle , X-Ray Diffraction , Photosynthesis/physiology , Glyceraldehyde-3-Phosphate Dehydrogenases/chemistry , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism
12.
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
13.
J Extracell Vesicles ; 10(9): e12114, 2021 07.
Article in English | MEDLINE | ID: mdl-34276899

ABSTRACT

Astrocytes-derived extracellular vesicles (EVs) are key players in glia-neuron communication. However, whether EVs interact with neurons at preferential sites and how EVs reach these sites on neurons remains elusive. Using optical manipulation to study single EV-neuron dynamics, we here show that large EVs scan the neuron surface and use neuronal processes as highways to move extracellularly. Large EV motion on neurites is driven by the binding of EV to a surface receptor that slides on neuronal membrane, thanks to actin cytoskeleton rearrangements. The use of prion protein (PrP)-coated synthetic beads and PrP knock out EVs/neurons points at vesicular PrP and its receptor(s) on neurons in the control of EV motion. Surprisingly, a fraction of large EVs contains actin filaments and has an independent capacity to move in an actin-mediated way, through intermittent contacts with the plasma membrane. Our results unveil, for the first time, a dual mechanism exploited by astrocytic large EVs to passively/actively reach target sites on neurons moving on the neuron surface.


Subject(s)
Astrocytes/cytology , Extracellular Vesicles/physiology , Neurites/physiology , Prion Proteins/metabolism , Actins/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Movement , Cells, Cultured , Cytoskeleton/physiology , Energy Metabolism , Female , Male , Rats , Rats, Sprague-Dawley , Surface Properties
14.
DNA Repair (Amst) ; 85: 102746, 2020 01.
Article in English | MEDLINE | ID: mdl-31739207

ABSTRACT

Nucleotide excision repair (NER) is a DNA repair pathway present in all domains of life. In bacteria, UvrA protein localizes the DNA lesion, followed by verification by UvrB helicase and excision by UvrC double nuclease. UvrA senses deformations and flexibility of the DNA duplex without precisely localizing the lesion in the damaged strand, an element essential for proper NER. Using a combination of techniques, we elucidate the mechanism of the damage verification step in bacterial NER. UvrA dimer recruits two UvrB molecules to its two sides. Each of the two UvrB molecules clamps a different DNA strand using its ß-hairpin element. Both UvrB molecules then translocate to the lesion, and UvrA dissociates. The UvrB molecule that clamps the damaged strand gets stalled at the lesion to recruit UvrC. This mechanism allows UvrB to verify the DNA damage and identify its precise location triggering subsequent steps in the NER pathway.


Subject(s)
Bacteria/genetics , DNA Helicases/chemistry , DNA Helicases/metabolism , Adenosine Triphosphatases/metabolism , Bacteria/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , DNA Damage , DNA Repair , Endodeoxyribonucleases/metabolism , Models, Molecular , Protein Conformation
15.
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
16.
Cells ; 8(9)2019 09 10.
Article in English | MEDLINE | ID: mdl-31510042

ABSTRACT

Bone marrow Mesenchymal Stem Cells (BM-MSCs), due to their strong protective and anti-inflammatory abilities, have been widely investigated in the context of several diseases for their possible therapeutic role, based on the release of a highly proactive secretome composed of soluble factors and Extracellular Vesicles (EVs). BM-MSC-EVs, in particular, convey many of the beneficial features of parental cells, including direct and indirect ß-amyloid degrading-activities, immunoregulatory and neurotrophic abilities. Therefore, EVs represent an extremely attractive tool for therapeutic purposes in neurodegenerative diseases, including Alzheimer's disease (AD). We examined the therapeutic potential of BM-MSC-EVs injected intracerebrally into the neocortex of APPswe/PS1dE9 AD mice at 3 and 5 months of age, a time window in which the cognitive behavioral phenotype is not yet detectable or has just started to appear. We demonstrate that BM-MSC-EVs are effective at reducing the Aß plaque burden and the amount of dystrophic neurites in both the cortex and hippocampus. The presence of Neprilysin on BM-MSC-EVs, opens the possibility of a direct ß-amyloid degrading action. Our results indicate a potential role for BM-MSC-EVs already in the early stages of AD, suggesting the possibility of intervening before overt clinical manifestations.


Subject(s)
Extracellular Vesicles/transplantation , Mesenchymal Stem Cells/metabolism , Plaque, Amyloid/therapy , Alzheimer Disease/genetics , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Cerebral Cortex/metabolism , Disease Models, Animal , Extracellular Vesicles/metabolism , Female , Hippocampus/metabolism , Male , Mesenchymal Stem Cell Transplantation/methods , Mice , Mice, Inbred C57BL , Neurites/metabolism
17.
Nat Commun ; 10(1): 1269, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30894521

ABSTRACT

Systemic light chain amyloidosis (AL)  is a life-threatening disease caused by aggregation and deposition of monoclonal immunoglobulin light chains (LC) in target organs. Severity of heart involvement is the most important factor determining prognosis. Here, we report the 4.0 Å resolution cryo-electron microscopy map and molecular model of amyloid fibrils extracted from the heart of an AL amyloidosis patient with severe amyloid cardiomyopathy. The helical fibrils are composed of a single protofilament, showing typical 4.9 Å stacking and cross-ß architecture. Two distinct polypeptide stretches (total of 77 residues) from the LC variable domain (Vl) fit the fibril density. Despite Vl high sequence variability, residues stabilizing the fibril core are conserved through different cardiotoxic Vl, highlighting structural motifs that may be common to misfolding-prone LCs. Our data shed light on the architecture of LC amyloids, correlate amino acid sequences with fibril assembly, providing the grounds for development of innovative medicines.


Subject(s)
Amyloid/ultrastructure , Immunoglobulin Light Chains/ultrastructure , Immunoglobulin Light-chain Amyloidosis/pathology , Myocardium/ultrastructure , Protein Aggregation, Pathological/pathology , Aged , Amino Acid Sequence , Amyloid/immunology , Amyloid/metabolism , Autopsy , Cryoelectron Microscopy , Humans , Immunoglobulin Light Chains/immunology , Immunoglobulin Light Chains/metabolism , Immunoglobulin Light-chain Amyloidosis/diagnosis , Immunoglobulin Light-chain Amyloidosis/immunology , Immunoglobulin Light-chain Amyloidosis/metabolism , Male , Myocardium/immunology , Myocardium/metabolism , Myocardium/pathology , Protein Aggregation, Pathological/diagnosis , Protein Aggregation, Pathological/immunology , Protein Aggregation, Pathological/metabolism , Protein Conformation, beta-Strand , Protein Folding , Sequence Alignment , Sequence Homology, Amino Acid , Severity of Illness Index
18.
Biophys Chem ; 225: 15-19, 2017 06.
Article in English | MEDLINE | ID: mdl-27989548

ABSTRACT

Eukaryotic DNA replication initiation and the Fanconi anemia pathway of interstrand crosslink repair both revolve around the recruitment of a set of DNA-processing factors onto a dimeric protein complex, which functions as a loading platform (MCM and FANCI-FANCD2 respectively). Here we compare and contrast the two systems, identifying a set of unresolved mechanistic questions. How is the dimeric loading platform assembled on the DNA? How can equivalent covalent modification of both factors in a dimer be achieved? Are multicomponent DNA-interacting machines built symmetrically around their dimeric loading platform? Recent biochemical reconstitution studies are starting to shed light on these issues.


Subject(s)
DNA Repair , DNA Replication , Animals , DNA/chemistry , Dimerization , Fanconi Anemia , Fanconi Anemia Complementation Group D2 Protein , Humans
19.
Cell Rep ; 18(3): 611-623, 2017 01 17.
Article in English | MEDLINE | ID: mdl-27986592

ABSTRACT

Activation of the main DNA interstrand crosslink repair pathway in higher eukaryotes requires mono-ubiquitination of FANCI and FANCD2 by FANCL, the E3 ligase subunit of the Fanconi anemia core complex. FANCI and FANCD2 form a stable complex; however, the molecular basis of their ubiquitination is ill defined. FANCD2 mono-ubiquitination by FANCL is stimulated by the presence of the FANCB and FAAP100 core complex components, through an unknown mechanism. How FANCI mono-ubiquitination is achieved remains unclear. Here, we use structural electron microscopy, combined with crosslink-coupled mass spectrometry, to find that FANCB, FANCL, and FAAP100 form a dimer of trimers, containing two FANCL molecules that are ideally poised to target both FANCI and FANCD2 for mono-ubiquitination. The FANCC-FANCE-FANCF subunits bridge between FANCB-FANCL-FAAP100 and the FANCI-FANCD2 substrate. A transient interaction with FANCC-FANCE-FANCF alters the FANCI-FANCD2 configuration, stabilizing the dimerization interface. Our data provide a model to explain how equivalent mono-ubiquitination of FANCI and FANCD2 occurs.


Subject(s)
Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Amino Acid Sequence , Chromatography, High Pressure Liquid , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dimerization , Fanconi Anemia Complementation Group D2 Protein/chemistry , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group Proteins/chemistry , Fanconi Anemia Complementation Group Proteins/genetics , Humans , Mass Spectrometry , Microscopy, Electron , Protein Multimerization , Protein Structure, Tertiary , Substrate Specificity , Ubiquitination
20.
Science ; 355(6320): 93-95, 2017 01 06.
Article in English | MEDLINE | ID: mdl-28059770

ABSTRACT

Retroviral integrase (IN) functions within the intasome nucleoprotein complex to catalyze insertion of viral DNA into cellular chromatin. Using cryo-electron microscopy, we now visualize the functional maedi-visna lentivirus intasome at 4.9 angstrom resolution. The intasome comprises a homo-hexadecamer of IN with a tetramer-of-tetramers architecture featuring eight structurally distinct types of IN protomers supporting two catalytically competent subunits. The conserved intasomal core, previously observed in simpler retroviral systems, is formed between two IN tetramers, with a pair of C-terminal domains from flanking tetramers completing the synaptic interface. Our results explain how HIV-1 IN, which self-associates into higher-order multimers, can form a functional intasome, reconcile the bulk of early HIV-1 IN biochemical and structural data, and provide a lentiviral platform for design of HIV-1 IN inhibitors.


Subject(s)
HIV Integrase/chemistry , HIV-1/chemistry , Virus Integration , Catalytic Domain , Cryoelectron Microscopy , DNA, Viral/chemistry , DNA, Viral/ultrastructure , Drug Design , HIV Integrase/ultrastructure , HIV Integrase Inhibitors/chemistry , HIV-1/enzymology , HIV-1/ultrastructure , Humans , Models, Molecular , Protein Domains , Static Electricity , Virus Assembly
SELECTION OF CITATIONS
SEARCH DETAIL