Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Autophagy ; 20(3): 557-576, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37938170

ABSTRACT

Macroautophagy/autophagy is a fundamental aspect of eukaryotic biology, and the autophagy-related protein ATG9A is part of the core machinery facilitating this process. In addition to ATG9A vertebrates encode ATG9B, a poorly characterized paralog expressed in a subset of tissues. Herein, we characterize the structure of human ATG9B revealing the conserved homotrimeric quaternary structure and explore the conformational dynamics of the protein. Consistent with the experimental structure and computational chemistry, we establish that ATG9B is a functional lipid scramblase. We show that ATG9B can compensate for the absence of ATG9A in starvation-induced autophagy displaying similar subcellular trafficking and steady-state localization. Finally, we demonstrate that ATG9B can form a heteromeric complex with ATG2A. By establishing the molecular structure and function of ATG9B, our results inform the exploration of niche roles for autophagy machinery in more complex eukaryotes and reveal insights relevant across species.Abbreviation: ATG: autophagy related; CHS: cholesteryl hemisuccinate; cryo-EM: single-particle cryogenic electron microscopy; CTF: contrast transfer function: CTH: C- terminal α helix; FSC: fourier shell correlation; HDIR: HORMA domain interacting region; LMNG: lauryl maltose neopentyl glycol; MD: molecular dynamics simulations; MSA: multiple sequence alignment; NBD-PE: 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine-N-(7-nitro-2-1,3-benzoxadiazol-4-yl ammonium salt); POPC: palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine; RBG: repeating beta groove domain; RMSD: root mean square deviation; SEC: size-exclusion chromatography; TMH: transmembrane helix.


Subject(s)
Autophagy , Membrane Proteins , Animals , Humans , Autophagy-Related Proteins/metabolism , Membrane Proteins/metabolism
2.
Cell ; 186(16): 3427-3442.e22, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37421949

ABSTRACT

SARS-CoV-2 is associated with broad tissue tropism, a characteristic often determined by the availability of entry receptors on host cells. Here, we show that TMEM106B, a lysosomal transmembrane protein, can serve as an alternative receptor for SARS-CoV-2 entry into angiotensin-converting enzyme 2 (ACE2)-negative cells. Spike substitution E484D increased TMEM106B binding, thereby enhancing TMEM106B-mediated entry. TMEM106B-specific monoclonal antibodies blocked SARS-CoV-2 infection, demonstrating a role of TMEM106B in viral entry. Using X-ray crystallography, cryogenic electron microscopy (cryo-EM), and hydrogen-deuterium exchange mass spectrometry (HDX-MS), we show that the luminal domain (LD) of TMEM106B engages the receptor-binding motif of SARS-CoV-2 spike. Finally, we show that TMEM106B promotes spike-mediated syncytium formation, suggesting a role of TMEM106B in viral fusion. Together, our findings identify an ACE2-independent SARS-CoV-2 infection mechanism that involves cooperative interactions with the receptors heparan sulfate and TMEM106B.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Receptors, Virus/metabolism , Virus Internalization , Protein Binding , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism
3.
Antimicrob Agents Chemother ; 67(7): e0046223, 2023 07 18.
Article in English | MEDLINE | ID: mdl-37310224

ABSTRACT

HIV-1 integrase-LEDGF allosteric inhibitors (INLAIs) share the binding site on the viral protein with the host factor LEDGF/p75. These small molecules act as molecular glues promoting hyper-multimerization of HIV-1 IN protein to severely perturb maturation of viral particles. Herein, we describe a new series of INLAIs based on a benzene scaffold that display antiviral activity in the single digit nanomolar range. Akin to other compounds of this class, the INLAIs predominantly inhibit the late stages of HIV-1 replication. A series of high-resolution crystal structures revealed how these small molecules engage the catalytic core and the C-terminal domains of HIV-1 IN. No antagonism was observed between our lead INLAI compound BDM-2 and a panel of 16 clinical antiretrovirals. Moreover, we show that compounds retained high antiviral activity against HIV-1 variants resistant to IN strand transfer inhibitors and other classes of antiretroviral drugs. The virologic profile of BDM-2 and the recently completed single ascending dose phase I trial (ClinicalTrials.gov identifier: NCT03634085) warrant further clinical investigation for use in combination with other antiretroviral drugs. Moreover, our results suggest routes for further improvement of this emerging drug class.


Subject(s)
HIV Infections , HIV Integrase Inhibitors , HIV Integrase , Humans , Virus Replication , HIV Integrase Inhibitors/pharmacology , HIV Integrase Inhibitors/therapeutic use , Antiviral Agents/pharmacology , HIV Integrase/metabolism , HIV Infections/drug therapy , Allosteric Regulation
4.
mBio ; 14(1): e0356022, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36744954

ABSTRACT

Allosteric HIV-1 integrase (IN) inhibitors (ALLINIs) are an emerging class of small molecules that disrupt viral maturation by inducing the aberrant multimerization of IN. Here, we present cocrystal structures of HIV-1 IN with two potent ALLINIs, namely, BI-D and the drug candidate Pirmitegravir. The structures reveal atomistic details of the ALLINI-induced interface between the HIV-1 IN catalytic core and carboxyl-terminal domains (CCD and CTD). Projecting from their principal binding pocket on the IN CCD dimer, the compounds act as molecular glue by engaging a triad of invariant HIV-1 IN CTD residues, namely, Tyr226, Trp235, and Lys266, to nucleate the CTD-CCD interaction. The drug-induced interface involves the CTD SH3-like fold and extends to the beginning of the IN carboxyl-terminal tail region. We show that mutations of HIV-1 IN CTD residues that participate in the interface with the CCD greatly reduce the IN-aggregation properties of Pirmitegravir. Our results explain the mechanism of the ALLINI-induced condensation of HIV-1 IN and provide a reliable template for the rational development of this series of antiretrovirals through the optimization of their key contacts with the viral target. IMPORTANCE Despite the remarkable success of combination antiretroviral therapy, HIV-1 remains among the major causes of human suffering and loss of life in poor and developing nations. To prevail in this drawn-out battle with the pandemic, it is essential to continue developing advanced antiviral agents to fight drug resistant HIV-1 variants. Allosteric integrase inhibitors (ALLINIs) are an emerging class of HIV-1 antagonists that are orthogonal to the current antiretroviral drugs. These small molecules act as highly specific molecular glue, which triggers the aggregation of HIV-1 integrase. In this work, we present high-resolution crystal structures that reveal the crucial interactions made by two potent ALLINIs, namely, BI-D and Pirmitegravir, with HIV-1 integrase. Our results explain the mechanism of drug action and will inform the development of this promising class of small molecules for future use in antiretroviral regimens.


Subject(s)
HIV Infections , HIV Integrase Inhibitors , Humans , Allosteric Regulation , HIV Integrase Inhibitors/pharmacology , Antiviral Agents/therapeutic use , HIV Infections/drug therapy
5.
Mol Cell ; 82(22): 4324-4339.e8, 2022 11 17.
Article in English | MEDLINE | ID: mdl-36347259

ABSTRACT

ATG9A and ATG2A are essential core members of the autophagy machinery. ATG9A is a lipid scramblase that allows equilibration of lipids across a membrane bilayer, whereas ATG2A facilitates lipid flow between tethered membranes. Although both have been functionally linked during the formation of autophagosomes, the molecular details and consequences of their interaction remain unclear. By combining data from peptide arrays, crosslinking, and hydrogen-deuterium exchange mass spectrometry together with cryoelectron microscopy, we propose a molecular model of the ATG9A-2A complex. Using this integrative structure modeling approach, we identify several interfaces mediating ATG9A-2A interaction that would allow a direct transfer of lipids from ATG2A into the lipid-binding perpendicular branch of ATG9A. Mutational analyses combined with functional activity assays demonstrate their importance for autophagy, thereby shedding light on this protein complex at the heart of autophagy.


Subject(s)
Autophagosomes , Autophagy , Cryoelectron Microscopy , Biological Assay , Lipids
6.
Cell Rep ; 40(8): 111276, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35981534

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike is the target for neutralizing antibodies elicited following both infection and vaccination. While extensive research has shown that the receptor binding domain (RBD) and, to a lesser extent, the N-terminal domain (NTD) are the predominant targets for neutralizing antibodies, identification of neutralizing epitopes beyond these regions is important for informing vaccine development and understanding antibody-mediated immune escape. Here, we identify a class of broadly neutralizing antibodies that bind an epitope on the spike subdomain 1 (SD1) and that have arisen from infection or vaccination. Using cryo-electron microscopy (cryo-EM) and hydrogen-deuterium exchange coupled to mass spectrometry (HDX-MS), we show that SD1-specific antibody P008_60 binds an epitope that is not accessible within the canonical prefusion states of the SARS-CoV-2 spike, suggesting a transient conformation of the viral glycoprotein that is vulnerable to neutralization.


Subject(s)
COVID-19 , SARS-CoV-2 , Antibodies, Neutralizing , Antibodies, Viral , Cryoelectron Microscopy , Epitopes , Humans , Neutralization Tests , Spike Glycoprotein, Coronavirus , Syndactyly , Vaccination
7.
Nat Commun ; 13(1): 2416, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35504909

ABSTRACT

A multimer of retroviral integrase (IN) synapses viral DNA ends within a stable intasome nucleoprotein complex for integration into a host cell genome. Reconstitution of the intasome from the maedi-visna virus (MVV), an ovine lentivirus, revealed a large assembly containing sixteen IN subunits1. Herein, we report cryo-EM structures of the lentiviral intasome prior to engagement of target DNA and following strand transfer, refined at 3.4 and 3.5 Å resolution, respectively. The structures elucidate details of the protein-protein and protein-DNA interfaces involved in lentiviral intasome formation. We show that the homomeric interfaces involved in IN hexadecamer formation and the α-helical configuration of the linker connecting the C-terminal and catalytic core domains are critical for MVV IN strand transfer activity in vitro and for virus infectivity. Single-molecule microscopy in conjunction with photobleaching reveals that the MVV intasome can bind a variable number, up to sixteen molecules, of the lentivirus-specific host factor LEDGF/p75. Concordantly, ablation of endogenous LEDGF/p75 results in gross redistribution of MVV integration sites in human and ovine cells. Our data confirm the importance of the expanded architecture observed in cryo-EM studies of lentiviral intasomes and suggest that this organization underlies multivalent interactions with chromatin for integration targeting to active genes.


Subject(s)
DNA, Viral , Integrases , Animals , Humans , Catalytic Domain , DNA, Viral/metabolism , Integrases/metabolism , Lentivirus/genetics , Lentivirus/metabolism , Models, Molecular , Retroviridae/genetics , Sheep/genetics , Virus Integration
9.
Nat Commun ; 12(1): 4996, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34404793

ABSTRACT

Between 10 and 20 million people worldwide are infected with the human T-cell lymphotropic virus type 1 (HTLV-1). Despite causing life-threatening pathologies there is no therapeutic regimen for this deltaretrovirus. Here, we screened a library of integrase strand transfer inhibitor (INSTI) candidates built around several chemical scaffolds to determine their effectiveness in limiting HTLV-1 infection. Naphthyridines with substituents in position 6 emerged as the most potent compounds against HTLV-1, with XZ450 having highest efficacy in vitro. Using single-particle cryo-electron microscopy we visualised XZ450 as well as the clinical HIV-1 INSTIs raltegravir and bictegravir bound to the active site of the deltaretroviral intasome. The structures reveal subtle differences in the coordination environment of the Mg2+ ion pair involved in the interaction with the INSTIs. Our results elucidate the binding of INSTIs to the HTLV-1 intasome and support their use for pre-exposure prophylaxis and possibly future treatment of HTLV-1 infection.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cryoelectron Microscopy , HTLV-I Infections/drug therapy , Human T-lymphotropic virus 1/drug effects , Amides , Catalytic Domain , Deltaretrovirus , Drug Resistance, Viral/drug effects , HIV Integrase/drug effects , HIV-1 , Heterocyclic Compounds, 3-Ring , Human T-lymphotropic virus 1/genetics , Humans , Naphthyridines/pharmacology , Piperazines , Pyridones , Recombinant Proteins
10.
Sci Adv ; 7(22)2021 05.
Article in English | MEDLINE | ID: mdl-33888467

ABSTRACT

The coronaviral spike is the dominant viral antigen and the target of neutralizing antibodies. We show that SARS-CoV-2 spike binds biliverdin and bilirubin, the tetrapyrrole products of heme metabolism, with nanomolar affinity. Using cryo-electron microscopy and x-ray crystallography, we mapped the tetrapyrrole interaction pocket to a deep cleft on the spike N-terminal domain (NTD). At physiological concentrations, biliverdin significantly dampened the reactivity of SARS-CoV-2 spike with immune sera and inhibited a subset of neutralizing antibodies. Access to the tetrapyrrole-sensitive epitope is gated by a flexible loop on the distal face of the NTD. Accompanied by profound conformational changes in the NTD, antibody binding requires relocation of the gating loop, which folds into the cleft vacated by the metabolite. Our results indicate that SARS-CoV-2 spike NTD harbors a dominant epitope, access to which can be controlled by an allosteric mechanism that is regulated through recruitment of a metabolite.


Subject(s)
COVID-19/immunology , Heme/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/immunology , Bilirubin/metabolism , Biliverdine/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , Epitopes , Humans , Immune Sera , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity
11.
ACS Infect Dis ; 7(6): 1469-1482, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33686850

ABSTRACT

Integrase strand transfer inhibitors (INSTIs) block the integration step of the retroviral lifecycle and are first-line drugs used for the treatment of HIV-1/AIDS. INSTIs have a polycyclic core with heteroatom triads, chelate the metal ions at the active site, and have a halobenzyl group that interacts with viral DNA attached to the core by a flexible linker. The most broadly effective INSTIs inhibit both wild-type (WT) integrase (IN) and a variety of well-known mutants. However, because there are mutations that reduce the potency of all of the available INSTIs, new and better compounds are needed. Models based on recent structures of HIV-1 and red-capped mangabey SIV INs suggest modifications in the INSTI structures that could enhance interactions with the 3'-terminal adenosine of the viral DNA, which could improve performance against INSTI resistant mutants. We designed and tested a series of INSTIs having modifications to their naphthyridine scaffold. One of the new compounds retained good potency against an expanded panel of HIV-1 IN mutants that we tested. Our results suggest the possibility of designing inhibitors that combine the best features of the existing compounds, which could provide additional efficacy against known HIV-1 IN mutants.


Subject(s)
HIV Integrase Inhibitors , HIV-1 , Pharmaceutical Preparations , Drug Resistance, Viral/genetics , HIV Integrase Inhibitors/pharmacology , HIV-1/genetics , Mutation
12.
medRxiv ; 2021 Jan 26.
Article in English | MEDLINE | ID: mdl-33532784

ABSTRACT

The coronaviral spike is the dominant viral antigen and the target of neutralizing antibodies. We show that SARS-CoV-2 spike binds biliverdin and bilirubin, the tetrapyrrole products of haem metabolism, with nanomolar affinity. Using cryo-electron microscopy and X-ray crystallography we mapped the tetrapyrrole interaction pocket to a deep cleft on the spike N-terminal domain (NTD). At physiological concentrations, biliverdin significantly dampened the reactivity of SARS-CoV-2 spike with immune sera and inhibited a subset of neutralizing antibodies. Access to the tetrapyrrole-sensitive epitope is gated by a flexible loop on the distal face of the NTD. Accompanied by profound conformational changes in the NTD, antibody binding requires relocation of the gating loop, which folds into the cleft vacated by the metabolite. Our results indicate that the virus co-opts the haem metabolite for the evasion of humoral immunity via allosteric shielding of a sensitive epitope and demonstrate the remarkable structural plasticity of the NTD.

13.
Nat Commun ; 11(1): 5043, 2020 10 07.
Article in English | MEDLINE | ID: mdl-33028863

ABSTRACT

Human T-cell lymphotropic virus type 1 (HTLV-1) is a deltaretrovirus and the most oncogenic pathogen. Many of the ~20 million HTLV-1 infected people will develop severe leukaemia or an ALS-like motor disease, unless a therapy becomes available. A key step in the establishment of infection is the integration of viral genetic material into the host genome, catalysed by the retroviral integrase (IN) enzyme. Here, we use X-ray crystallography and single-particle cryo-electron microscopy to determine the structure of the functional deltaretroviral IN assembled on viral DNA ends and bound to the B56γ subunit of its human host factor, protein phosphatase 2 A. The structure reveals a tetrameric IN assembly bound to two molecules of the phosphatase via a conserved short linear motif. Insight into the deltaretroviral intasome and its interaction with the host will be crucial for understanding the pattern of integration events in infected individuals and therefore bears important clinical implications.


Subject(s)
Human T-lymphotropic virus 1/pathogenicity , Integrases/ultrastructure , Protein Phosphatase 2/ultrastructure , Simian T-lymphotropic virus 1/enzymology , Viral Proteins/ultrastructure , Virus Integration , Amino Acid Motifs/genetics , Cloning, Molecular , Cryoelectron Microscopy , Crystallography, X-Ray , DNA, Viral/metabolism , DNA, Viral/ultrastructure , Human T-lymphotropic virus 1/enzymology , Human T-lymphotropic virus 1/genetics , Humans , Integrases/genetics , Integrases/metabolism , Leukemia-Lymphoma, Adult T-Cell/pathology , Leukemia-Lymphoma, Adult T-Cell/virology , Molecular Docking Simulation , Mutagenesis, Site-Directed , Paraparesis, Tropical Spastic/pathology , Paraparesis, Tropical Spastic/virology , Protein Multimerization , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Protein Structure, Quaternary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Sequence Homology, Amino Acid , Simian T-lymphotropic virus 1/genetics , Single Molecule Imaging , Viral Proteins/genetics , Viral Proteins/metabolism
14.
Structure ; 28(8): 954-962.e4, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32521228

ABSTRACT

CDC7 is an essential Ser/Thr kinase that acts upon the replicative helicase throughout the S phase of the cell cycle and is activated by DBF4. Here, we present crystal structures of a highly active human CDC7-DBF4 construct. The structures reveal a zinc-finger domain at the end of the kinase insert 2 that pins the CDC7 activation loop to motif M of DBF4 and the C lobe of CDC7. These interactions lead to ordering of the substrate-binding platform and full opening of the kinase active site. In a co-crystal structure with a mimic of MCM2 Ser40 phosphorylation target, the invariant CDC7 residues Arg373 and Arg380 engage phospho-Ser41 at substrate P+1 position, explaining the selectivity of the S-phase kinase for Ser/Thr residues followed by a pre-phosphorylated or an acidic residue. Our results clarify the role of DBF4 in activation of CDC7 and elucidate the structural basis for recognition of its preferred substrates.


Subject(s)
Cell Cycle Proteins/chemistry , Protein Serine-Threonine Kinases/chemistry , Catalytic Domain , Cell Cycle Proteins/metabolism , Humans , Molecular Docking Simulation , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Substrate Specificity , Zinc Fingers
15.
Nat Struct Mol Biol ; 27(1): 78-83, 2020 01.
Article in English | MEDLINE | ID: mdl-31907454

ABSTRACT

The human integral membrane protein SERINC5 potently restricts HIV-1 infectivity and sensitizes the virus to antibody-mediated neutralization. Here, using cryo-EM, we determine the structures of human SERINC5 and its orthologue from Drosophila melanogaster at subnanometer and near-atomic resolution, respectively. The structures reveal a novel fold comprised of ten transmembrane helices organized into two subdomains and bisected by a long diagonal helix. A lipid binding groove and clusters of conserved residues highlight potential functional sites. A structure-based mutagenesis scan identified surface-exposed regions and the interface between the subdomains of SERINC5 as critical for HIV-1-restriction activity. The same regions are also important for viral sensitization to neutralizing antibodies, directly linking the antiviral activity of SERINC5 with remodeling of the HIV-1 envelope glycoprotein.


Subject(s)
HIV Infections/immunology , HIV-1/immunology , Membrane Proteins/chemistry , Membrane Proteins/immunology , Animals , Drosophila Proteins/chemistry , Drosophila Proteins/ultrastructure , Drosophila melanogaster/chemistry , Humans , Membrane Proteins/ultrastructure , Models, Molecular , Protein Conformation , Protein Domains , Protein Multimerization
16.
Nat Commun ; 10(1): 4189, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31519882

ABSTRACT

Retroviral integrase can efficiently utilise nucleosomes for insertion of the reverse-transcribed viral DNA. In face of the structural constraints imposed by the nucleosomal structure, integrase gains access to the scissile phosphodiester bonds by lifting DNA off the histone octamer at the site of integration. To clarify the mechanism of DNA looping by integrase, we determined a 3.9 Å resolution structure of the prototype foamy virus intasome engaged with a nucleosome core particle. The structural data along with complementary single-molecule Förster resonance energy transfer measurements reveal twisting and sliding of the nucleosomal DNA arm proximal to the integration site. Sliding the nucleosomal DNA by approximately two base pairs along the histone octamer accommodates the necessary DNA lifting from the histone H2A-H2B subunits to allow engagement with the intasome. Thus, retroviral integration into nucleosomes involves the looping-and-sliding mechanism for nucleosomal DNA repositioning, bearing unexpected similarities to chromatin remodelers.


Subject(s)
Cryoelectron Microscopy/methods , DNA/chemistry , Histones/chemistry , Nucleosomes/metabolism , DNA/ultrastructure , Fluorescence Resonance Energy Transfer , Histones/ultrastructure , Humans , Nucleoproteins/chemistry , Nucleoproteins/ultrastructure , Nucleosomes/ultrastructure , Protein Structure, Quaternary
17.
Nucleic Acids Res ; 47(9): 4663-4683, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30916345

ABSTRACT

Cleavage factor I mammalian (CFIm) complex, composed of cleavage and polyadenylation specificity factor 5 (CPSF5) and serine/arginine-like protein CPSF6, regulates alternative polyadenylation (APA). Loss of CFIm function results in proximal polyadenylation site usage, shortening mRNA 3' untranslated regions (UTRs). Although CPSF6 plays additional roles in human disease, its nuclear translocation mechanism remains unresolved. Two ß-karyopherins, transportin (TNPO) 1 and TNPO3, can bind CPSF6 in vitro, and we demonstrate here that while the TNPO1 binding site is dispensable for CPSF6 nuclear import, the arginine/serine (RS)-like domain (RSLD) that mediates TNPO3 binding is critical. The crystal structure of the RSLD-TNPO3 complex revealed potential CPSF6 interaction residues, which were confirmed to mediate TNPO3 binding and CPSF6 nuclear import. Both binding and nuclear import were independent of RSLD phosphorylation, though a hyperphosphorylated mimetic mutant failed to bind TNPO3 and mislocalized to the cell cytoplasm. Although hypophosphorylated CPSF6 largely supported normal polyadenylation site usage, a significant number of mRNAs harbored unnaturally extended 3' UTRs, similar to what is observed when other APA regulators, such as CFIIm component proteins, are depleted. Our results clarify the mechanism of CPSF6 nuclear import and highlight differential roles for RSLD phosphorylation in nuclear translocation versus regulation of APA.


Subject(s)
Polyadenylation/genetics , Protein Conformation , RNA-Binding Proteins/chemistry , beta Karyopherins/chemistry , Active Transport, Cell Nucleus/genetics , Crystallography, X-Ray , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Humans , Phosphorylation/genetics , Protein Binding/genetics , Protein Serine-Threonine Kinases/chemistry , Protein-Tyrosine Kinases/chemistry , RNA, Messenger , RNA-Binding Proteins/genetics , beta Karyopherins/genetics , mRNA Cleavage and Polyadenylation Factors/chemistry , mRNA Cleavage and Polyadenylation Factors/genetics
18.
Mol Cell ; 72(1): 112-126.e5, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30217558

ABSTRACT

Maintenance of epigenetic integrity relies on coordinated recycling and partitioning of parental histones and deposition of newly synthesized histones during DNA replication. This process depends upon a poorly characterized network of histone chaperones, remodelers, and binding proteins. Here we implicate the POLE3-POLE4 subcomplex of the leading-strand polymerase, Polε, in replication-coupled nucleosome assembly through its ability to selectively bind to histones H3-H4. Using hydrogen/deuterium exchange mass spectrometry and physical mapping, we define minimal domains necessary for interaction between POLE3-POLE4 and histones H3-H4. Biochemical analyses establish that POLE3-POLE4 is a histone chaperone that promotes tetrasome formation and DNA supercoiling in vitro. In cells, POLE3-POLE4 binds both newly synthesized and parental histones, and its depletion hinders helicase unwinding and chromatin PCNA unloading and compromises coordinated parental histone retention and new histone deposition. Collectively, our study reveals that POLE3-POLE4 possesses intrinsic H3-H4 chaperone activity, which facilitates faithful nucleosome dynamics at the replication fork.


Subject(s)
DNA Polymerase III/genetics , DNA Replication/genetics , DNA-Binding Proteins/genetics , Epigenesis, Genetic/genetics , Histones/biosynthesis , Nucleoproteins/genetics , Chromatin/genetics , DNA Polymerase II/chemistry , DNA Polymerase II/genetics , DNA Polymerase III/chemistry , DNA-Binding Proteins/chemistry , Histone Chaperones/chemistry , Histone Chaperones/genetics , Histones/genetics , Humans , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Nucleoproteins/chemistry , Nucleosomes/chemistry , Nucleosomes/genetics , Poly-ADP-Ribose Binding Proteins/chemistry , Poly-ADP-Ribose Binding Proteins/genetics , Proliferating Cell Nuclear Antigen/genetics , Protein Binding
19.
Nat Commun ; 8(1): 2241, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29269875

ABSTRACT

Eukaryotic origins of replication are licensed upon loading of the MCM helicase motor onto DNA. ATP hydrolysis by MCM is required for loading and the post-catalytic MCM is an inactive double hexamer that encircles duplex DNA. Origin firing depends on MCM engagement of Cdc45 and GINS to form the CMG holo-helicase. CMG assembly requires several steps including MCM phosphorylation by DDK. To understand origin activation, here we have determined the cryo-EM structures of DNA-bound MCM, either unmodified or phosphorylated, and visualize a phospho-dependent MCM element likely important for Cdc45 recruitment. MCM pore loops touch both the Watson and Crick strands, constraining duplex DNA in a bent configuration. By comparing our new MCM-DNA structure with the structure of CMG-DNA, we suggest how the conformational transition from the loaded, post-catalytic MCM to CMG might promote DNA untwisting and melting at the onset of replication.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Replication , DNA-Binding Proteins/ultrastructure , DNA/ultrastructure , Minichromosome Maintenance Proteins/ultrastructure , Nuclear Proteins/ultrastructure , Nucleic Acid Conformation , Protein Conformation , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/ultrastructure , Cryoelectron Microscopy , DNA/metabolism , DNA Helicases , DNA-Binding Proteins/metabolism , Holoenzymes , Image Processing, Computer-Assisted , Minichromosome Maintenance Proteins/metabolism , Nuclear Proteins/metabolism , Phosphorylation , Protein Structure, Quaternary , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism
20.
J Med Chem ; 60(17): 7315-7332, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28737946

ABSTRACT

Integrase mutations can reduce the effectiveness of the first-generation FDA-approved integrase strand transfer inhibitors (INSTIs), raltegravir (RAL) and elvitegravir (EVG). The second-generation agent, dolutegravir (DTG), has enjoyed considerable clinical success; however, resistance-causing mutations that diminish the efficacy of DTG have appeared. Our current findings support and extend the substrate envelope concept that broadly effective INSTIs can be designed by filling the envelope defined by the DNA substrates. Previously, we explored 1-hydroxy-2-oxo-1,2-dihydro-1,8-naphthyridine-3-carboxamides as an INSTI scaffold, making a limited set of derivatives, and concluded that broadly effective INSTIs can be developed using this scaffold. Herein, we report an extended investigation of 6-substituents as well the first examples of 7-substituted analogues of this scaffold. While 7-substituents are not well-tolerated, we have identified novel substituents at the 6-position that are highly effective, with the best compound (6p) retaining better efficacy against a broad panel of known INSTI resistant mutants than any analogues we have previously described.


Subject(s)
HIV Integrase Inhibitors/chemistry , HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , HIV-1/drug effects , Naphthyridines/chemistry , Naphthyridines/pharmacology , Cell Line , Crystallography, X-Ray , Drug Resistance, Viral , HIV Infections/drug therapy , HIV Infections/virology , HIV Integrase/chemistry , HIV Integrase/genetics , HIV-1/enzymology , HIV-1/genetics , HIV-1/physiology , Humans , Models, Molecular , Mutation , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...