28 results on '"Paolo Swuec"'
Search Results
2. Astrocytes‐derived extracellular vesicles in motion at the neuron surface: Involvement of the prion protein
- Author
-
Giulia D'Arrigo, Martina Gabrielli, Federica Scaroni, Paolo Swuec, Ladan Amin, Anna Pegoraro, Elena Adinolfi, Francesco Di Virgilio, Dan Cojoc, Giuseppe Legname, and Claudia Verderio
- Subjects
astrocytes ,cytoskeleton ,extracellular vesicles ,neurons ,optical tweezers ,prion protein ,Cytology ,QH573-671 - Abstract
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.
- Published
- 2021
- Full Text
- View/download PDF
3. The FA Core Complex Contains a Homo-dimeric Catalytic Module for the Symmetric Mono-ubiquitination of FANCI-FANCD2
- Author
-
Paolo Swuec, Ludovic Renault, Aaron Borg, Fenil Shah, Vincent J. Murphy, Sylvie van Twest, Ambrosius P. Snijders, Andrew J. Deans, and Alessandro Costa
- Subjects
interstrand crosslink ,Fanconi anemia ,ubiquitin ligase ,DNA repair ,cryo-electron microscopy ,mono-ubiquitination ,Biology (General) ,QH301-705.5 - 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.
- Published
- 2017
- Full Text
- View/download PDF
4. Intracerebral Injection of Extracellular Vesicles from Mesenchymal Stem Cells Exerts Reduced Aβ Plaque Burden in Early Stages of a Preclinical Model of Alzheimer’s Disease
- Author
-
Chiara A. Elia, Matteo Tamborini, Marco Rasile, Genni Desiato, Sara Marchetti, Paolo Swuec, Sonia Mazzitelli, Francesca Clemente, Achille Anselmo, Michela Matteoli, Maria Luisa Malosio, and Silvia Coco
- Subjects
bone marrow mesenchymal stem cells ,extracellular vesicles ,Alzheimer’s disease ,APPswe/PS1dE9 AD mice ,Neprilysin ,dystrophic neuritis ,SMI ,Aβ plaques ,Cytology ,QH573-671 - 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 β-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 Aβ 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 β-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.
- Published
- 2019
- Full Text
- View/download PDF
5. DNA binding polarity, dimerization, and ATPase ring remodeling in the CMG helicase of the eukaryotic replisome
- Author
-
Alessandro Costa, Ludovic Renault, Paolo Swuec, Tatjana Petojevic, James J Pesavento, Ivar Ilves, Kirsty MacLellan-Gibson, Roland A Fleck, Michael R Botchan, and James M Berger
- Subjects
DNA replication ,Mcm2-7 ,helicase ,motor protein ,replication fork ,AAA+ ATPase ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The Cdc45/Mcm2-7/GINS (CMG) helicase separates DNA strands during replication in eukaryotes. How the CMG is assembled and engages DNA substrates remains unclear. Using electron microscopy, we have determined the structure of the CMG in the presence of ATPγS and a DNA duplex bearing a 3′ single-stranded tail. The structure shows that the MCM subunits of the CMG bind preferentially to single-stranded DNA, establishes the polarity by which DNA enters into the Mcm2-7 pore, and explains how Cdc45 helps prevent DNA from dissociating from the helicase. The Mcm2-7 subcomplex forms a cracked-ring, right-handed spiral when DNA and nucleotide are bound, revealing unexpected congruencies between the CMG and both bacterial DnaB helicases and the AAA+ motor of the eukaryotic proteasome. The existence of a subpopulation of dimeric CMGs establishes the subunit register of Mcm2-7 double hexamers and together with the spiral form highlights how Mcm2-7 transitions through different conformational and assembly states as it matures into a functional helicase.
- Published
- 2014
- Full Text
- View/download PDF
6. THE RIBOREGULATION MECHANISM OF HUMAN SERINE HYDROXYMETHYLTRANSFERASE IS ROOTED IN AN ALLOSTERIC SWITCH
- Author
-
Sharon Spizzichino, Federica Di Fonzo, Chiara Marabelli, Angela Tramonti, Antonio Chaves-Sanjuan, Alessia Parroni, Giovanna Boumis, Francesca Romana Liberati, Alessio Paone, Linda Celeste Montemiglio, Matteo Ardini, Arjen J. Jakobi, Alok Bharadwaj, Paolo Swuec, Gian Gaetano Tartaglia, Alessandro Paiardini, Roberto Contestabile, Serena Rinaldo, Martino Bolognesi, Giorgio Giardina, and Francesca Cutruzzolà
- Abstract
SUMMARYRNA can directly control protein activity in a process called riboregulation; only a few mechanisms of riboregulation have been described in detail, none of these being characterized on structural grounds. Here we present a comprehensive structural, functional, and phylogenetic analysis of riboregulation of cytosolic serine hydroxymethyltransferase (SHMT1), the enzyme interconverting serine and glycine in one-carbon metabolism. We show that the RNA modulator competes with polyglutamylated folates and acts as an allosteric switch, selectively altering the enzyme’s reactivity vs. serine. In addition, we identify the tetrameric assembly and a flap structural motif as key structural elements necessary for binding of RNA to eukaryotic SHMT1. The results presented here suggest that riboregulation may have played a role in the evolution of eukaryotic SHMT1 and the compartmentalization of one-carbon metabolism. The findings also provide insights for RNA-based therapeutic strategies targeting this cancer-linked metabolic pathway.
- Published
- 2023
- Full Text
- View/download PDF
7. The USR domain of USF1 mediates NF-Y interactions and cooperative DNA binding
- Author
-
Dana Saad, Nerina Gnesutta, Erica Valentini, Roberto Mantovani, Antonio Chaves-Sanjuan, Melissa A. Graewert, Andrea Bernardini, Petr V. Konarev, Marco Nardini, Matteo Pigni, Paolo Swuec, Dmitri I. Svergun, and Mariangela Lorenzo
- Subjects
genetic structures ,biology ,Chemistry ,USF1 ,Colocalization ,Promoter ,DNA ,General Medicine ,DNA-binding domain ,Biochemistry ,Cell biology ,chemistry.chemical_compound ,Gene Expression Regulation ,Protein Domains ,Structural Biology ,Gene expression ,biology.protein ,Humans ,Upstream Stimulatory Factors ,Promoter Regions, Genetic ,Molecular Biology ,Peptide sequence ,Transcription factor ,Protein Binding - 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.
- Published
- 2021
- Full Text
- View/download PDF
8. Mass spectrometry characterization of light chain fragmentation sites in cardiac AL amyloidosis: insights into the timing of proteolysis
- Author
-
Antonio Chaves-Sanjuan, Masayoshi Tasaki, Paola Rognoni, Mario Nuvolone, Stefano Ricagno, Serena Caminito, Paolo Swuec, Paolo Milani, Giampaolo Merlini, Federica Iavarone, Francesca Lavatelli, Giovanni Palladini, Andrea Urbani, and Giulia Mazzini
- Subjects
0301 basic medicine ,Amyloid ,proteolysis ,Genomics and Proteomics ,Proteolysis ,Protein aggregation ,Immunoglobulin light chain ,Fibril ,Biochemistry ,Protein Structure, Secondary ,protein aggregation ,amyloid fibrils ,03 medical and health sciences ,proteomics ,Protein structure ,Tandem Mass Spectrometry ,protein conformation ,medicine ,Humans ,structural biology ,Electrophoresis, Gel, Two-Dimensional ,Immunoglobulin Light-chain Amyloidosis ,mass spectrometry (MS) ,Amino Acid Sequence ,protein structure ,Molecular Biology ,Chromatography, High Pressure Liquid ,030102 biochemistry & molecular biology ,medicine.diagnostic_test ,fibril ,Chemistry ,Myocardium ,Amyloidosis ,Cell Biology ,medicine.disease ,Protein Structure, Tertiary ,030104 developmental biology ,Structural biology ,Protein Structure and Folding ,Biophysics ,Immunoglobulin Light Chains ,Protein folding ,Peptides ,cardiomyopathy - 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.
- Published
- 2020
- Full Text
- View/download PDF
9. Cryo-EM Structures of Azospirillum brasilense Glutamate Synthase in Its Oligomeric Assemblies
- Author
-
Martino Bolognesi, Carlo Camilloni, Paolo Swuec, Antonio Chaves-Sanjuan, and Vanoni
- Subjects
Iron-Sulfur Proteins ,biology ,Flavin Mononucleotide ,Stereochemistry ,Chemistry ,Cryo-electron microscopy ,Cryoelectron Microscopy ,Glutamate Synthase ,Flavoprotein ,Azospirillum brasilense ,biology.organism_classification ,Catalysis ,Cofactor ,Electron Transport ,Electron transfer ,Structural Biology ,Glutamate synthase ,Flavin-Adenine Dinucleotide ,biology.protein ,Molecule ,Molecular Biology - 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 β-subunit (βGltS) that assemble in different αβ oligomers in solution. Here, we present the cryo-electron microscopy structures of Azospirillum brasilense GltS in four different oligomeric states (α4β3, α4β4, α6β4 and α6β6, in the 3.5- to 4.1-A 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 βGltS.
- Published
- 2019
- Full Text
- View/download PDF
10. Cryo-EM structure of cardiac amyloid fibrils from an immunoglobulin light chain AL amyloidosis patient
- Author
-
Giovanni Palladini, Martino Bolognesi, Francesca Lavatelli, Stefano Ricagno, Paolo Milani, Paola Rognoni, Pierluigi Mauri, Masayoshi Tasaki, Martina Maritan, Francesca Brambilla, Giampaolo Merlini, Cristina Paissoni, Paolo Swuec, and Carlo Camilloni
- Subjects
Proteomics ,Male ,Amyloid ,Protein Folding ,Cryo-electron microscopy ,Science ,macromolecular substances ,Immunoglobulin light chain ,Fibril ,Protein Aggregation, Pathological ,Severity of Illness Index ,Article ,medicine ,AL amyloidosis ,Humans ,Immunoglobulin Light-chain Amyloidosis ,Amino Acid Sequence ,Structural motif ,lcsh:Science ,Cryo-EM ,Aged ,Sequence Homology, Amino Acid ,Chemistry ,Amyloidosis ,Myocardium ,Cryoelectron Microscopy ,medicine.disease ,Biophysics ,lcsh:Q ,Immunoglobulin Light Chains ,Protein Conformation, beta-Strand ,Autopsy ,Amyloid cardiomyopathy ,Sequence Alignment - 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-β 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., Immunoglobulin Light Chain Amyloidosis (AL) is the most common systemic amyloidosis occurring in Western countries. Here the authors present the 4.0 Å cryo-EM structure of light chain AL55 fibrils that were isolated from the heart of an AL systemic amyloidosis patient.
- Published
- 2019
- Full Text
- View/download PDF
11. Structural clues of Serine hidroxymethyltransferase (SHMT) riboregulation
- Author
-
Spizzichino, Sharon, Chiara, Marabelli, Antonio Chaves Sanjuan, Paolo, Swuec, Montemiglio, LINDA CELESTE, Matteo, Ardini, Rinaldo, Serena, Paone, Alessio, Boumis, Giovanna, Giardina, Giorgio, Martino, Bolognesi, and Cutruzzola', Francesca
- Published
- 2021
12. High-Light Versus Low-Light: Effects on Paired Photosystem II Supercomplex Structural Rearrangement in Pea Plants
- Author
-
Roberto Marotta, Cristina Pagliano, Giuseppe Zanotti, Pascal Albanese, Guido Saracco, Martino Bolognesi, Paolo Swuec, and Alessandro Grinzato
- Subjects
0106 biological sciences ,0301 basic medicine ,Light ,Photosystem II ,Cryo-electron microscopy ,Light-Harvesting Protein Complexes ,Stacking ,cryo-electron microscopy ,macromolecular substances ,Thylakoids ,01 natural sciences ,Article ,Catalysis ,Inorganic Chemistry ,lcsh:Chemistry ,03 medical and health sciences ,light acclimation ,photosystem II supercomplex ,Physical and Theoretical Chemistry ,Molecular Biology ,lcsh:QH301-705.5 ,Spectroscopy ,plant thylakoid membranes ,Chemistry ,Organic Chemistry ,Peas ,Photosystem II Protein Complex ,food and beverages ,General Medicine ,Cryo‐electron microscopy ,Light effect ,Computer Science Applications ,030104 developmental biology ,lcsh:Biology (General) ,lcsh:QD1-999 ,Photoprotection ,Thylakoid ,Biophysics ,Light acclimation ,Photosystem II supercomplex ,Plant thylakoid membranes ,Variable number ,010606 plant biology & botany - 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.
- Published
- 2020
13. A combined structural and biochemical approach reveals translocation and stalling of UvrB on the DNA lesion as a mechanism of damage verification in bacterial nucleotide excision repair
- Author
-
Marcin Jaciuk, Marcin Nowotny, Paolo Swuec, Ludovic Renault, Vineet Gaur, Shivlee Nirwal, Janusz M. Bujnicki, Alessandro Costa, Mateusz Dobrychłop, and Joanna M. Kasprzak
- Subjects
Models, Molecular ,DNA Repair ,Protein Conformation ,DNA repair ,DNA damage ,Biochemistry & Proteomics ,Biochemistry ,Imaging ,Lesion ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Bacterial Proteins ,medicine ,A-DNA ,Molecular Biology ,030304 developmental biology ,Adenosine Triphosphatases ,0303 health sciences ,Nuclease ,Endodeoxyribonucleases ,Bacteria ,biology ,Genome Integrity & Repair ,DNA Helicases ,Helicase ,Cell Biology ,Cell biology ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,bacteria ,Cell Cycle & Chromosomes ,medicine.symptom ,DNA ,DNA Damage ,Nucleotide excision repair ,Structural Biology & Biophysics - 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 β-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.
- Published
- 2020
- Full Text
- View/download PDF
14. Cas9 Allosteric Inhibition by the Anti-CRISPR Protein AcrIIA6
- Author
-
Christian Cambillau, Béatrice Amigues, Geneviève M. Rousseau, Sylvain Moineau, Claire Zimberger, Alain Roussel, Olivier Fuchsbauer, Antonio Chaves-Sanjuan, Yannick Doyon, Alexis Duringer, Adeline Goulet, Sébastien Lévesque, Minja Velimirovic, Paolo Swuec, Silvia Spinelli, Daniel Agudelo, Martino Bolognesi, Architecture et fonction des macromolécules biologiques (AFMB), Aix Marseille Université (AMU)-Centre National de la Recherche Scientifique (CNRS)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Università degli Studi di Milano [Milano] (UNIMI), Centre de recherche du CHU de Québec-Université Laval (CRCHUQ), CHU de Québec–Université Laval, Université Laval [Québec] (ULaval)-Université Laval [Québec] (ULaval), Faculté de médecine dentaire [Université Laval, Québec], Université Laval [Québec] (ULaval), Dpt de Microbiologie-Infectiologie et d’Immunologie [Laval], Department of Biosciences [Milano], Félix d'Hérelle Reference Center for Bacterial Viruses, Groupe de Recherche en Écologie Buccale (GREB), Centre National de la Recherche Scientifique (CNRS), Aix Marseille Université (AMU), NSERC of Canada, Canada RGPIN-2014-05132/RGPIN-2014-05698Fonds de la Recherche en Sante du Quebec FRQS 254294United States Department of Health & Human ServicesNational Institutes of Health (NIH) - USA R01-GM129325/P41-GM103311, ANR-18-CE11-0016,PHARE,Vengeance de phages: analyses structurales et fonctionnelles de protéines anti CRISPR-Cas9(2018), ANR-10-INBS-0005,FRISBI,Infrastructure Française pour la Biologie Structurale Intégrée(2010), Hôpital de l'Enfant-Jésus [CHU Québec] (HEJ), Faculté de médecine de l'Université Laval [Québec] (ULaval), ANR-10-INSB-05-01 INSB ANR-10-INSB-0501, Fonds de la recherche du Québec-Santé (FRQS 254294)NIH R01-GM129325 and P41-GM103311, Discovery program, RGPIN-2014-05132, RGPIN-2014-05698, Università degli Studi di Milano = University of Milan (UNIMI), Laboratoire d'ingénierie des systèmes macromoléculaires (LISM), and Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
bacteriophages ,Protein Conformation ,[SDV]Life Sciences [q-bio] ,Allosteric regulation ,anti-CRISPR protein ,Virulence ,cryo-electron microscopy ,Biology ,Genome ,Structure-Activity Relationship ,Viral Proteins ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Allosteric Regulation ,CRISPR-Associated Protein 9 ,Hydrolase ,Escherichia coli ,Humans ,Streptococcus thermophilus ,CRISPR ,Clustered Regularly Interspaced Short Palindromic Repeats ,Streptococcus thermophilus Cas9 ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Binding Sites ,Cas9 ,DNA ,Cell Biology ,biology.organism_classification ,3. Good health ,Cell biology ,St1Cas9 ,Kinetics ,chemistry ,Mutation ,CRISPR-Cas Systems ,CRISPR-Cas9 ,K562 Cells ,030217 neurology & neurosurgery ,Bacteria ,Protein Binding - Abstract
International audience; Molecular CellArticleCas9 Allosteric Inhibitionby the Anti-CRISPR Protein AcrIIA6Olivier Fuchsbauer,1,2,9Paolo Swuec,3,4,9Claire Zimberger,1,2Be ́atrice Amigues,1,2Se ́bastien Levesque,5Daniel Agudelo,5Alexis Duringer,5Antonio Chaves-Sanjuan,4Silvia Spinelli,1,2Genevie`ve M. Rousseau,6,7Minja Velimirovic,5Martino Bolognesi,3,4Alain Roussel,1,2Christian Cambillau,1,2Sylvain Moineau,6,7,8Yannick Doyon,5and Adeline Goulet1,2,10,*1Architecture et Fonction des Macromole ́cules Biologiques, Centre National de la Recherche Scientifique (CNRS), Campus de Luminy, Case932, 13288 Marseille Cedex 09, France2Architecture et Fonction des Macromole ́cules Biologiques, Aix-Marseille Universite ́, Campus de Luminy, Case 932, 13288 Marseille Cedex09, France3Dipartimento di Bioscienze, Universita`degli Studi di Milano, Via Celoria 26, 20133 Milano, Italy4Centro di Ricerca Pediatrica Romeo ed Enrica Invernizzi, Universita`degli Studi di Milano, Via Celoria 26, 20133 Milano, Italy5Centre Hospitalier Universitaire de Que ́bec–Universite ́Laval Research Center, Que ́bec City, QC G1V 4G2, Canada6De ́partement de biochimie, de microbiologie, et de bio-informatique, Faculte ́des sciences et de ge ́nie, Universite ́Laval, Que ́bec City, QC,G1V 0A6, Canada7Groupe de recherche en e ́cologie buccale, Faculte ́de me ́decine dentaire, Universite ́Laval, Que ́bec City, QC, G1V 0A6, Canada8Fe ́lix d’He ́relle Reference Center for Bacterial Viruses, Faculte ́de me ́decine dentaire, Universite ́Laval, Que ́bec City, QC, G1V 0A6, Canada9These authors contributed equally10Lead Contact*Correspondence:adeline.goulet@afmb.univ-mrs.frhttps://doi.org/10.1016/j.molcel.2019.09.012SUMMARYIn the arms race against bacteria, bacteriophageshave evolved diverse anti-CRISPR proteins (Acrs)that block CRISPR-Cas immunity. Acrs play key rolesin the molecular coevolution of bacteria with theirpredators, use a variety of mechanisms of action,and provide tools to regulate Cas-based genomemanipulation. Here, we present structural and func-tional analyses of AcrIIA6, an Acr from virulentphages, exploring its unique anti-CRISPR action.Our cryo-EM structures and functional data ofAcrIIA6 binding toStreptococcus thermophilusCas9 (St1Cas9) show that AcrIIA6 acts as an allo-steric inhibitor and induces St1Cas9 dimerization.AcrIIA6 reduces St1Cas9 binding affinity for DNAand prevents DNA binding within cells. The PAMand AcrIIA6 recognition sites are structurally closeand allosterically linked. Mechanistically, AcrIIA6 af-fects the St1Cas9 conformational dynamics associ-ated with PAM binding. Finally, we identify a naturalSt1Cas9 variant resistant to AcrIIA6 illustratingAcr-driven mutational escape and molecular diversi-fication of Cas9 proteins
- Published
- 2019
- Full Text
- View/download PDF
15. Intracerebral Injection of Extracellular Vesicles from Mesenchymal Stem Cells Exerts Reduced Aβ Plaque Burden in Early Stages of a Preclinical Model of Alzheimer's Disease
- Author
-
Genni Desiato, Maria Luisa Malosio, Matteo Tamborini, Marco Rasile, Sonia Mazzitelli, Paolo Swuec, Achille Anselmo, Chiara A. Elia, Silvia Coco, Sara Marchetti, Michela Matteoli, Francesca Clemente, Elia, C, Tamborini, M, Rasile, M, Desiato, G, Marchetti, S, Swuec, P, Mazzitelli, S, Clemente, F, Anselmo, A, Matteoli, M, Luisa Malosio, M, and Coco, S
- Subjects
Male ,Hippocampus ,Context (language use) ,Plaque, Amyloid ,Disease ,Aβ plaques ,Mesenchymal Stem Cell Transplantation ,Article ,Extracellular Vesicles ,Mice ,Alzheimer Disease ,ddc:570 ,Neurites ,Medicine ,Animals ,lcsh:QH301-705.5 ,Neprilysin ,Cerebral Cortex ,Neocortex ,Amyloid beta-Peptides ,SMI ,biology ,business.industry ,dystrophic neuritis ,Mesenchymal stem cell ,Brain ,bone marrow mesenchymal stem cells ,dystrophic neuriti ,Mesenchymal Stem Cells ,General Medicine ,Cortex (botany) ,APPswe/PS1dE9 AD mice ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,lcsh:Biology (General) ,bone marrow mesenchymal stem cell ,biology.protein ,Female ,extracellular vesicle ,business ,Neuroscience ,Alzheimer’s disease ,Neurotrophin - 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 &beta, amyloid degrading-activities, immunoregulatory and neurotrophic abilities. Therefore, EVs represent an extremely attractive tool for therapeutic purposes in neurodegenerative diseases, including Alzheimer&rsquo, 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 A&beta, 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 &beta, 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.
- Published
- 2019
- Full Text
- View/download PDF
16. Gating movements and ion permeation in HCN4 pacemaker channels
- Author
-
Anna Moroni, Wayne A. Hendrickson, Giacomo Parisi, Dario DiFrancesco, Martino Bolognesi, Federica Gasparri, Atiyeh Sadat Sharifzadeh, Bina Santoro, Alessandro Porro, Antonio Chaves-Sanjuan, Oliver B. Clarke, Filippo Mancia, Paolo Swuec, Andrea Saponaro, Gabriele Cerutti, M. Hunter Giese, Henry M. Colecraft, Daniel Bauer, Laura Alberio, Gerhard Thiel, Kay Hamacher, and Steven A. Siegelbaum
- Subjects
Cell Membrane Permeability ,Potassium Channels ,Cryo-electron microscopy ,Muscle Proteins ,Gating ,Article ,metal ion ,Cell Line ,03 medical and health sciences ,Molecular dynamics ,0302 clinical medicine ,cAMP ,Cyclic AMP ,Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels ,HCN channel ,Humans ,HCN4 ,HCN4 pore gating selectivity permeation cAMP metal ion cryo-EM molecular dynamics pacemaker ,Molecular Biology ,030304 developmental biology ,Ions ,0303 health sciences ,biology ,Chemistry ,Cryoelectron Microscopy ,selectivity ,Cell Biology ,Permeation ,molecular dynamics ,pacemaker ,Transmembrane domain ,Cytosol ,HEK293 Cells ,gating ,Biophysics ,biology.protein ,cryo-EM ,permeation ,Selectivity ,Ion Channel Gating ,pore ,Linker ,030217 neurology & neurosurgery - Abstract
Summary 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., Graphical abstract, Highlights • HCN4 structure is shown in ligand-free and ligand-bound state • Pore domain is shown in closed and in open configuration • Permeability and selectivity mechanisms of HCN channels are uncovered • A metal ion coordination site functionally couples cytoplasmic and transmembrane domains, HCN4 channels underlie the pacemaker current that controls heart rate. Saponaro et al. report the structure of HCN4 with the pore in closed and in open configuration and provide information on ion permeability and selectivity. In HCN4, a metal ion coordination site functionally connects the C-linker to the S4-S5 linker.
- Published
- 2021
- Full Text
- View/download PDF
17. The FA Core Complex Contains a Homo-dimeric Catalytic Module for the Symmetric Mono-ubiquitination of FANCI-FANCD2
- Author
-
Fenil Shah, Andrew J. Deans, Aaron J. Borg, Ludovic Renault, Alessandro Costa, Paolo Swuec, Sylvie van Twest, Vincent J. Murphy, and Ambrosius P. Snijders
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,DNA repair ,Protein subunit ,cryo-electron microscopy ,interstrand crosslink ,ubiquitin ligase ,General Biochemistry, Genetics and Molecular Biology ,Article ,Mass Spectrometry ,Substrate Specificity ,03 medical and health sciences ,Protein structure ,Fanconi anemia ,hemic and lymphatic diseases ,FANCD2 ,medicine ,Humans ,FANCL ,Amino Acid Sequence ,lcsh:QH301-705.5 ,Chromatography, High Pressure Liquid ,biology ,Chemistry ,Fanconi Anemia Complementation Group D2 Protein ,Ubiquitination ,nutritional and metabolic diseases ,medicine.disease ,mono-ubiquitination ,Fanconi Anemia Complementation Group Proteins ,3. Good health ,FANCB ,Ubiquitin ligase ,Protein Structure, Tertiary ,DNA-Binding Proteins ,Microscopy, Electron ,030104 developmental biology ,lcsh:Biology (General) ,Biochemistry ,Biophysics ,biology.protein ,Protein Multimerization ,Dimerization - Abstract
Summary 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., Graphical Abstract, Highlights • FANCB, FANCL, and FAAP100 form a symmetric dimer of trimers • FANCL is ideally poised for the symmetric mono-ubiquitination of FANCI-FANCD2 • Two separate FANCC-FANCE-FANCF complexes bind to the opposing poles of FANCB-FANCL-FAAP100 • FANCC-FANCE-FANCF stabilizes FANCI-FANCD2 for efficient mono-ubiquitination, Mono-ubiquitination of FANCI-FANCD2 by the Fanconi anemia core complex activates a major DNA interstrand-crosslink repair pathway important for genome stability maintenance. Here, Swuec et al. reveal the structural basis of this reaction by showing that the core complex exists as a dimeric catalytic module for the symmetric mono-ubiquitination of FANCI-FANCD2.
- Published
- 2016
18. Extracellular Vesicles from Mesenchymal Stem Cells reduce Aβ plaque burden in early stages of Alzheimer's disease
- Author
-
Chiara A. Elia, Matteo Tamborini, Marco Rasile, Genni Desiato, Paolo Swuec, Sara Marchetti, Sonia Mazzitelli, Francesca Clemente, Achille Anselmo, Michela Matteoli, Maria Luisa Malosio, Silvia Coco, Elia, C, Tamborini, M, Rasile, M, Desiato, G, Swuec, P, Marchetti, S, Mazzitelli, S, Clemente, F, Anselmo, A, Matteoli, M, Luisa Malosio, M, and Coco, S
- Subjects
SMI ,bone marrow mesenchymal stem cell ,Neprilysin ,dystrophic neuriti ,extracellular vesicle ,Aβ plaques ,Alzheimer’s disease ,APPswe/PS1dE9 AD mice - Abstract
Object Bone marrow mesenchymal stem cells (MSC), due to their strong protective and anti-inflammatory abilities, are 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). MSC-EVs, in particular, convey many of the beneficial features of parental cells, including direct and indirect β-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 intracerebrally injected MSC-EVs into the neocortex of APP/PS1 mice at 3 and 5 months of age, a time window in which the cognitive behavioral phenotype is not yet detectable or just starts to appear. Materials Primary cultures of Bone Marrow-derived MSC (up to P12 passage), APPswe/PS1dE9 (APP/PS1) AD mice. Methods MSC were stimulated by serum-deprivation for 3 hrs and supernatant was submitted to differential ultracentrifugation to collect EVs (including exosomes and microvesicles pools), which were characterized by Nanoparticle tracking, cryo-EM, flow cytometry and Western blot analysis. APP/PS1 mice, 3 and 5 months old, were injected intracortically with 4 uL of BM-MSC-derived EV suspension, corresponding to 22.4 ug of total proteins. Brain sections of mice treated or not with EVs were immunohistochemically stained for Abeta1-42 peptide (6-E10 antibody). Smi31 and 32 antibodies recognizing Neurofilaments were used for staining dystrophic neurites around Abeta1-42 plaques stained by Thioflavin-T. Results Intracerebral injection of MSC-EVs into the neocortex of APP/PS1 mice at 3 and 5 months of age reduced Abeta1-42 plaques burden one month later compared to same-age untreated mice. At 3 months, when plaques have just started to form, treatment conferred a preventive significance. In addition, following treatment with MSC-EVs, a reduction in dystrophic neurites could been measured. This decrease resulted significantly different in 6 month-old AD mice. Neprilysin, a metal-membrane endopeptidase able to degrade Abeta1-42, was detected on MSC-derived EV lysates. Discussion We demonstrate that MSC-EVs are effective in reducing the Aβ plaque burden and the amount of dystrophic neurites, in both cortex and hippocampus. The presence of Neprilysin on MSC-EVs opens the possibility of a direct β-amyloid degrading-action as a possible mechanisms of action. Conclusions Our results indicate a potential role for MSC-EVs already at early stages of AD, suggesting the possibility to intervene before overt clinical manifestations.
- Published
- 2019
19. Cas9 Allosteric Inhibition by the Anti-CRISPR Protein AcrIIA6
- Author
-
Geneviève M. Rousseau, Alexis Duringer, Claire Zimberger, Olivier Fuchsbauer, Adeline Goulet, Martino Bolognesi, Antonio Chaves-Sanjuan, Béatrice Amigues, Alain Roussel, Yannick Doyon, Sébastien Lévesque, Paolo Swuec, Silvia Spinelli, Daniel Agudelo, Christian Cambillau, and Sylvain Moineau
- Subjects
chemistry.chemical_compound ,Streptococcus thermophilus ,chemistry ,biology ,Cas9 ,Allosteric regulation ,Virulence ,CRISPR ,biology.organism_classification ,Genome ,DNA ,Bacteria ,Cell biology - 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 productive 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.
- Published
- 2019
- Full Text
- View/download PDF
20. Identification of a Small Molecule That Compromises the Structural Integrity of Viroplasms and Rotavirus Double-Layered Particles
- Author
-
Francesca Arnoldi, Matteo de Rosa, Michela Bollati, Eloise Mastrangelo, Paolo Swuec, Guido Papa, Oscar R. Burrone, Catherine Eichwald, Elisabeth M. Schraner, Mathias Ackermann, Giuditta De Lorenzo, Mario Milani, University of Zurich, Arnoldi, Francesca, Eichwald, Catherine, De Lorenzo, Giuditta, Schraner, Elisabeth M., Papa, Guido, Bollati, Michela, Swuec, Paolo, de Rosa, Matteo, Milani, Mario, Mastrangelo, Eloise, Ackermann, Mathia, and Burrone, Oscar R.
- Subjects
0301 basic medicine ,Rotavirus ,RNA polymerase III ,1109 Insect Science ,10077 Institute of Veterinary Anatomy ,medicine.disease_cause ,Virus Replication ,Transcription (biology) ,Chlorocebus aethiops ,Sf9 Cells ,Enzyme Inhibitors ,Viroplasm ,ML-60218 ,2404 Microbiology ,Antivirals ,VP6 ,DLP ,Drug ,Viral genome replication ,10244 Institute of Virology ,Inhibitor ,Viral protein ,Immunology ,Biology ,Microbiology ,Virus ,Cell Line ,Small Molecule Libraries ,03 medical and health sciences ,Viral Proteins ,Virology ,Vaccines and Antiviral Agents ,medicine ,Viroplasms ,Insect Science ,Animals ,Antiviral ,Viral Structures ,2403 Immunology ,Dose-Response Relationship, Drug ,RNA ,RNA Polymerase III ,Rotaviru ,030104 developmental biology ,Viral replication ,2406 Virology ,570 Life sciences ,biology - 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.
- Published
- 2017
- Full Text
- View/download PDF
21. A supramolecular assembly mediates lentiviral DNA integration
- Author
-
Julia Locke, Valgerdur Andrésdóttir, Erik Serrao, Ian A. Taylor, Stefán R. Jónsson, Nicola J. Cook, Peter Cherepanov, Alan Engelman, Valerie E. Pye, Paolo Swuec, Abhay Kotecha, Alessandro Costa, Daniel P. Maskell, and Allison Ballandras-Colas
- Subjects
0301 basic medicine ,Models, Molecular ,General Science & Technology ,Virus Integration ,Static Electricity ,PROTEIN ,Nanotechnology ,HIV Integrase ,HIV-1 INTEGRASE ,Article ,Supramolecular assembly ,03 medical and health sciences ,INTASOME ,DOMAIN ,Protein Domains ,Catalytic Domain ,BINDING ,Humans ,CRYSTAL-STRUCTURE ,Angstrom ,DNA Integration ,HIV Integrase Inhibitors ,Dna viral ,Multidisciplinary ,Science & Technology ,030102 biochemistry & molecular biology ,biology ,Virus Assembly ,Cryoelectron Microscopy ,IN-VITRO ,Integrase ,Cell biology ,Chromatin ,Nucleoprotein ,Multidisciplinary Sciences ,MODEL ,030104 developmental biology ,Drug Design ,DNA, Viral ,biology.protein ,HIV-1 ,Science & Technology - Other Topics ,RETROVIRAL INTEGRATION - Abstract
High-resolution insights into the intasome An essential step in the life cycle of lentiviruses such as HIV-1 is when viral DNA integrates into the host genome, establishing a permanent infection of the host cell. The viral integrase enzyme catalyzes this process and is a major drug target. During viral integration, integrase binds the ends of viral DNA, forming a higher-order structure called the intasome. Passos et al. and Ballandras-Colas et al. used cryo—electron microscopy to solve the structures of the intasomes from HIV-1 and maedi-visna virus (ovine lentivirus), respectively. These structures reveal how integrase self-associates to form a functional intasome and help resolve previous conflicting models of intasome assembly. Science , this issue p. 89 , p. 93
- Published
- 2016
22. DNA replication and inter-strand crosslink repair: Symmetric activation of dimeric nanomachines?
- Author
-
Paolo Swuec and Alessandro Costa
- Subjects
0301 basic medicine ,DNA Replication ,DNA Repair ,DNA repair ,Dimer ,Biophysics ,Covalent modification ,Eukaryotic DNA replication ,Interstrand crosslink ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,hemic and lymphatic diseases ,Animals ,Humans ,Chemistry ,Fanconi Anemia Complementation Group D2 Protein ,Organic Chemistry ,DNA replication ,DNA ,030104 developmental biology ,Fanconi Anemia ,Dimerization - 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.
- Published
- 2016
23. Human RECQ1 helicase-driven DNA unwinding, annealing, and branch migration: insights from DNA complex structures
- Author
-
Ying Zhang, Ashley C. W. Pike, Alessandro Costa, Francesca Marino, Alessandro Vindigni, Shivasankari Gomathinayagam, Frank von Delft, Paolo Swuec, Ludovic Renault, Opher Gileadi, Christina Schnecke, and Matteo Berti
- Subjects
Insecta ,Molecular Conformation ,DNA, Single-Stranded ,Protein dimer ,DNA helicases ,Plasma protein binding ,Crystallography, X-Ray ,Nucleic Acid Denaturation ,fork reversal ,chemistry.chemical_compound ,RecQ ,Escherichia coli ,Holliday junction ,Animals ,Humans ,Genetics ,DNA, Cruciform ,Multidisciplinary ,RecQ Helicases ,biology ,Nucleotides ,DNA replication ,Helicase ,DNA ,Biological Sciences ,Branch migration ,Protein Structure, Tertiary ,enzymes and coenzymes (carbohydrates) ,Zinc ,chemistry ,Chromatography, Gel ,biology.protein ,Biophysics ,Crystallization ,genome stability ,Protein Binding - 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.
- Published
- 2015
24. DNA binding polarity, dimerization, and ATPase ring remodeling in the CMG helicase of the eukaryotic replisome
- Author
-
Ludovic Renault, Roland A. Fleck, Kirsty MacLellan-Gibson, James J. Pesavento, Ivar Ilves, Michael R. Botchan, James M. Berger, Alessandro Costa, Tatjana Petojevic, and Paolo Swuec
- Subjects
Models, Molecular ,Chromosomal Proteins, Non-Histone ,Cell Cycle Proteins ,Biochemistry ,chemistry.chemical_compound ,Adenosine Triphosphate ,Single-Stranded ,Models ,Drosophila Proteins ,Biology (General) ,Adenosine Triphosphatases ,0303 health sciences ,Microscopy ,D. melanogaster ,biology ,Minichromosome Maintenance Proteins ,General Neuroscience ,030302 biochemistry & molecular biology ,RNA-Binding Proteins ,General Medicine ,Biophysics and Structural Biology ,Mcm2-7 ,DNA-Binding Proteins ,Chromosomal Proteins ,helicase ,Eukaryotic Cells ,Drosophila melanogaster ,Medicine ,RNA Splicing Factors ,AAA+ ATPase ,Research Article ,Protein Binding ,DNA Replication ,Protein Structure ,QH301-705.5 ,Science ,DNA, Single-Stranded ,DNA replication ,Electron ,General Biochemistry, Genetics and Molecular Biology ,Quaternary ,03 medical and health sciences ,motor protein ,Genetics ,Animals ,Protein Structure, Quaternary ,dnaB helicase ,030304 developmental biology ,CMG complex ,General Immunology and Microbiology ,Circular bacterial chromosome ,motor proteins ,Helicase ,Molecular ,Non-Histone ,DNA ,GINS ,Protein Structure, Tertiary ,Repressor Proteins ,Microscopy, Electron ,Protein Subunits ,chemistry ,Multiprotein Complexes ,biology.protein ,Biophysics ,Replisome ,replication fork ,Biochemistry and Cell Biology ,Protein Multimerization ,Tertiary - Abstract
The Cdc45/Mcm2-7/GINS (CMG) helicase separates DNA strands during replication in eukaryotes. How the CMG is assembled and engages DNA substrates remains unclear. Using electron microscopy, we have determined the structure of the CMG in the presence of ATPγS and a DNA duplex bearing a 3′ single-stranded tail. The structure shows that the MCM subunits of the CMG bind preferentially to single-stranded DNA, establishes the polarity by which DNA enters into the Mcm2-7 pore, and explains how Cdc45 helps prevent DNA from dissociating from the helicase. The Mcm2-7 subcomplex forms a cracked-ring, right-handed spiral when DNA and nucleotide are bound, revealing unexpected congruencies between the CMG and both bacterial DnaB helicases and the AAA+ motor of the eukaryotic proteasome. The existence of a subpopulation of dimeric CMGs establishes the subunit register of Mcm2-7 double hexamers and together with the spiral form highlights how Mcm2-7 transitions through different conformational and assembly states as it matures into a functional helicase. DOI: http://dx.doi.org/10.7554/eLife.03273.001, eLife digest Before a cell divides, it must duplicate its DNA so that each new cell inherits its own copy of the genome. To do this, the DNA double helix must be unwound so that the two individual strands of DNA can serve as templates for making new DNA molecules. Unwinding begins when two helicase complexes, termed the Mcm2-7 rings, are loaded together onto the DNA. At first, the two Mcm2-7 rings encircle the double-stranded DNA and remain bound together in an inactive form. Activating the Mcm2-7 rings requires the binding of five other proteins to each ring, which forms two larger complexes called CMG helicases. When the CMG helicases form, the two DNA strands separate and an individual Mcm2-7 ring ends up encircling each of the single DNA strands. However, how an activated CMG complex is assembled, and how it binds to and unwinds DNA, is not fully understood. Now, Costa et al. have determined the three-dimensional structure of the fruit fly CMG helicase bound to a DNA double helix with a single-stranded overhang at one end. The activated Mcm2-7 ring binds to the overhang, which confirms previous findings indicating that the activated helicase prefers single-stranded over double-stranded DNA. The structure also shows that, as a CMG helicase slides along the single-stranded DNA towards the double-stranded DNA, it is the ring complex's ‘motor domains’ that lead the way, while its DNA-binding domains trail behind. Costa et al. also found that disrupting some of the interactions between two of the five proteins that bind to the Mcm2-7 ring either prevented the replicative helicase from forming or made it unstable. Furthermore, it was revealed that one of these two proteins—called Cdc45—was ideally placed to capture the strand of DNA that might be accidentally released from the Mcm2-7 ring. It was also discovered that when the complex is bound to DNA, the motor domains of the Mcm2-7 complex change shape from a flat ring to a spiral structure; the DNA-binding domains, however, remain in a flat ring. Costa et al. note that this structure is similar to that adopted by many viral and bacterial helicases, and that it even shares many features with the molecular machinery that breaks down unneeded or damaged proteins inside cells. Finally, Costa et al. were able to image a structure composed of two CMG complexes bound together. This reveals the relative orientation of the two Mcm2-7 rings before they separate and move in opposite directions to unravel the DNA. The findings of Costa et al., combined with previous structural work in this field, demonstrate that the Mcm2-7 helicase complex can adopt many different shapes as it is assembled on DNA and activated to support DNA replication. DOI: http://dx.doi.org/10.7554/eLife.03273.002
- Published
- 2014
25. Author response: DNA binding polarity, dimerization, and ATPase ring remodeling in the CMG helicase of the eukaryotic replisome
- Author
-
James J. Pesavento, Roland A. Fleck, Paolo Swuec, James M. Berger, Michael R. Botchan, Tatjana Petojevic, Ivar Ilves, Kirsty MacLellan-Gibson, Ludovic Renault, and Alessandro Costa
- Subjects
chemistry.chemical_compound ,biology ,chemistry ,Polarity (physics) ,ATPase ,Biophysics ,biology.protein ,Replisome ,Helicase ,Ring (chemistry) ,DNA - Published
- 2014
- Full Text
- View/download PDF
26. Architecture and DNA Recognition Elements of the Fanconi Anemia FANCM-FAAP24 Complex
- Author
-
Maureen Bowles, Andrew J. Deans, Rachel Coulthard, Neil Q. McDonald, Stephen C. West, Alessandro Costa, and Paolo Swuec
- Subjects
Models, Molecular ,congenital, hereditary, and neonatal diseases and abnormalities ,HMG-box ,DNA Repair ,DNA repair ,DNA damage ,Cell Survival ,Mitomycin ,Oligonucleotides ,Biology ,Crystallography, X-Ray ,DNA-binding protein ,Article ,Protein Structure, Secondary ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Adenosine Triphosphate ,Fanconi anemia ,Coordination Complexes ,Structural Biology ,hemic and lymphatic diseases ,Catalytic Domain ,medicine ,Humans ,FANCM ,RNA, Small Interfering ,Molecular Biology ,030304 developmental biology ,Genetics ,0303 health sciences ,Oligonucleotide ,Hydrolysis ,Cryoelectron Microscopy ,DNA Helicases ,nutritional and metabolic diseases ,medicine.disease ,Fanconi Anemia Complementation Group Proteins ,DNA-Binding Proteins ,HEK293 Cells ,chemistry ,030220 oncology & carcinogenesis ,Gene Knockdown Techniques ,Nucleic Acid Conformation ,DNA ,DNA Damage ,Mutagens ,Protein Binding - Abstract
Summary Fanconi anemia (FA) is a disorder associated with a failure in DNA repair. FANCM (defective in FA complementation group M) and its partner FAAP24 target other FA proteins to sites of DNA damage. FANCM-FAAP24 is related to XPF/MUS81 endonucleases but lacks endonucleolytic activity. We report a structure of an FANCM C-terminal fragment (FANCMCTD) bound to FAAP24 and DNA. This S-shaped structure reveals the FANCM (HhH)2 domain is buried, whereas the FAAP24 (HhH)2 domain engages DNA. We identify a second DNA contact and a metal center within the FANCM pseudo-nuclease domain and demonstrate that mutations in either region impair double-stranded DNA binding in vitro and FANCM-FAAP24 function in vivo. We show the FANCM translocase domain lies in proximity to FANCMCTD by electron microscopy and that binding fork DNA structures stimulate its ATPase activity. This suggests a tracking model for FANCM-FAAP24 until an encounter with a stalled replication fork triggers ATPase-mediated fork remodeling., Highlights • FANCM-FAAP24 contains a large head and a curved hook tail • ATPase activity of FANCM translocase domain is stimulated by forked DNA structures • An S-shaped FANCM-FAAP24 fragment binds DNA through FAAP24 hairpins • Both FANCM PND and FAAP24 hairpins are required for FANCM function in vivo, The FANCM-FAAP24 complex anchors the Fanconi anemia core complex onto chromatin and activates DNA repair. Coulthard et al. describe the FANCM-FAAP24 architecture and characterize multiple DNA-binding sites. Forked DNA structures stimulate FANCM ATPase activity, which is important for stalled replication fork remodelling.
- Published
- 2013
- Full Text
- View/download PDF
27. Prediction of inhibitory activities of Hsp90 inhibitors
- Author
-
Paolo Swuec and David J. Barlow
- Subjects
Binding Sites ,Artificial neural network ,biology ,Chemistry ,Organic Chemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Crystallographic data ,Quantitative Structure-Activity Relationship ,Ligands ,Biochemistry ,Hsp90 ,Backpropagation ,Protein Structure, Tertiary ,Drug Discovery ,biology.protein ,Molecular Medicine ,Humans ,Thermodynamics ,Computer Simulation ,HSP90 Heat-Shock Proteins ,Biological system ,Molecular Biology ,Software - 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.
- Published
- 2011
28. Molecular mechanism of double Holliday junction dissolution
- Author
-
Alessandro Costa and Paolo Swuec
- Subjects
Genetics ,biology ,Helicase ,Review ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,chemistry.chemical_compound ,enzymes and coenzymes (carbohydrates) ,chemistry ,Holliday junction ,Biophysics ,biology.protein ,medicine ,Translocase ,Topoisomerase III ,Homologous recombination ,Function (biology) ,DNA ,Werner syndrome - Abstract
Processing of homologous recombination intermediates is tightly coordinated to ensure that chromosomal integrity is maintained and tumorigenesis avoided. Decatenation of double Holliday junctions, for example, is catalysed by two enzymes that work in tight coordination and belong to the same ‘dissolvasome’ complex. Within the dissolvasome, the RecQ-like BLM helicase provides the translocase function for Holliday junction migration, while the topoisomerase III alpha-RMI1 subcomplex works as a proficient DNA decatenase, together resulting in double-Holliday-junction unlinking. Here, we review the available architectural and biochemical knowledge on the dissolvasome machinery, with a focus on the structural interplay between its components.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.