Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 4155, 2023 07 12.
Article in English | MEDLINE | ID: mdl-37438348

ABSTRACT

The small Ultra-Red Fluorescent Protein (smURFP) represents a new class of fluorescent protein with exceptional photostability and brightness derived from allophycocyanin in a previous directed evolution. Here, we report the smURFP crystal structure to better understand properties and enable further engineering of improved variants. We compare this structure to the structures of allophycocyanin and smURFP mutants to identify the structural origins of the molecular brightness. We then use a structure-guided approach to develop monomeric smURFP variants that fluoresce with phycocyanobilin but not biliverdin. Furthermore, we measure smURFP photophysical properties necessary for advanced imaging modalities, such as those relevant for two-photon, fluorescence lifetime, and single-molecule imaging. We observe that smURFP has the largest two-photon cross-section measured for a fluorescent protein, and that it produces more photons than organic dyes. Altogether, this study expands our understanding of the smURFP, which will inform future engineering toward optimal FPs compatible with whole organism studies.


Subject(s)
Biliverdine , Coloring Agents , Luminescent Proteins/genetics , Engineering , Red Fluorescent Protein
2.
Sci Adv ; 9(7): eadg2997, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36791199

ABSTRACT

The assembly of the autophagy initiation machinery nucleates autophagosome biogenesis, including in the PINK1- and Parkin-dependent mitophagy pathway implicated in Parkinson's disease. The structural interaction between the sole transmembrane autophagy protein, autophagy-related protein 9A (ATG9A), and components of the Unc-51-like autophagy activating kinase (ULK1) complex is one of the major missing links needed to complete a structural map of autophagy initiation. We determined the 2.4-Å x-ray crystallographic structure of the ternary structure of ATG9A carboxyl-terminal tail bound to the ATG13:ATG101 Hop1/Rev7/Mad2 (HORMA) dimer, which is part of the ULK1 complex. We term the interacting portion of the extreme carboxyl-terminal part of the ATG9A tail the "HORMA dimer-interacting region" (HDIR). This structure shows that the HDIR binds to the HORMA domain of ATG101 by ß sheet complementation such that the ATG9A tail resides in a deep cleft at the ATG13:ATG101 interface. Disruption of this complex in cells impairs damage-induced PINK1/Parkin mitophagy mediated by the cargo receptor NDP52.


Subject(s)
Membrane Proteins , Mitophagy , Autophagy-Related Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Protein Kinases/genetics
3.
Elife ; 102021 09 10.
Article in English | MEDLINE | ID: mdl-34505572

ABSTRACT

Autophagy is a cellular process that degrades cytoplasmic cargo by engulfing it in a double-membrane vesicle, known as the autophagosome, and delivering it to the lysosome. The ATG12-5-16L1 complex is responsible for conjugating members of the ubiquitin-like ATG8 protein family to phosphatidylethanolamine in the growing autophagosomal membrane, known as the phagophore. ATG12-5-16L1 is recruited to the phagophore by a subset of the phosphatidylinositol 3-phosphate-binding seven-bladedß -propeller WIPI proteins. We determined the crystal structure of WIPI2d in complex with the WIPI2 interacting region (W2IR) of ATG16L1 comprising residues 207-230 at 1.85 Å resolution. The structure shows that the ATG16L1 W2IR adopts an alpha helical conformation and binds in an electropositive and hydrophobic groove between WIPI2 ß-propeller blades 2 and 3. Mutation of residues at the interface reduces or blocks the recruitment of ATG12-5-16 L1 and the conjugation of the ATG8 protein LC3B to synthetic membranes. Interface mutants show a decrease in starvation-induced autophagy. Comparisons across the four human WIPIs suggest that WIPI1 and 2 belong to a W2IR-binding subclass responsible for localizing ATG12-5-16 L1 and driving ATG8 lipidation, whilst WIPI3 and 4 belong to a second W34IR-binding subclass responsible for localizing ATG2, and so directing lipid supply to the nascent phagophore. The structure provides a framework for understanding the regulatory node connecting two central events in autophagy initiation, the action of the autophagic PI 3-kinase complex on the one hand and ATG8 lipidation on the other.


Subject(s)
Autophagosomes/metabolism , Autophagy-Related Proteins/metabolism , Autophagy , Intracellular Membranes/metabolism , Membrane Proteins/metabolism , Phosphate-Binding Proteins/metabolism , Autophagosomes/genetics , Autophagy-Related Protein 8 Family/metabolism , Autophagy-Related Proteins/chemistry , Autophagy-Related Proteins/genetics , Crystallography , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Membrane Proteins/chemistry , Membrane Proteins/genetics , Models, Molecular , Phosphate-Binding Proteins/chemistry , Phosphate-Binding Proteins/genetics , Phosphatidylinositol 3-Kinase/metabolism , Point Mutation , Protein Conformation, alpha-Helical , Protein Transport , Signal Transduction , Structure-Activity Relationship
4.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article in English | MEDLINE | ID: mdl-33361333

ABSTRACT

The molecular basis for the severity and rapid spread of the COVID-19 disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is largely unknown. ORF8 is a rapidly evolving accessory protein that has been proposed to interfere with immune responses. The crystal structure of SARS-CoV-2 ORF8 was determined at 2.04-Šresolution by X-ray crystallography. The structure reveals a ∼60-residue core similar to SARS-CoV-2 ORF7a, with the addition of two dimerization interfaces unique to SARS-CoV-2 ORF8. A covalent disulfide-linked dimer is formed through an N-terminal sequence specific to SARS-CoV-2, while a separate noncovalent interface is formed by another SARS-CoV-2-specific sequence, 73YIDI76 Together, the presence of these interfaces shows how SARS-CoV-2 ORF8 can form unique large-scale assemblies not possible for SARS-CoV, potentially mediating unique immune suppression and evasion activities.


Subject(s)
Molecular Structure , SARS-CoV-2/chemistry , Viral Proteins/chemistry , Evolution, Molecular , Immune Evasion
5.
bioRxiv ; 2020 Aug 27.
Article in English | MEDLINE | ID: mdl-32869027

ABSTRACT

The molecular basis for the severity and rapid spread of the COVID-19 disease caused by SARS-CoV-2 is largely unknown. ORF8 is a rapidly evolving accessory protein that has been proposed to interfere with immune responses. The crystal structure of SARS-CoV-2 ORF8 was determined at 2.04 Å resolution by x-ray crystallography. The structure reveals a ~60 residue core similar to SARS-CoV ORF7a with the addition of two dimerization interfaces unique to SARS-CoV-2 ORF8. A covalent disulfide-linked dimer is formed through an N-terminal sequence specific to SARS-CoV-2, while a separate non-covalent interface is formed by another SARS-CoV-2-specific sequence, 73 YIDI 76 . Together the presence of these interfaces shows how SARS-CoV-2 ORF8 can form unique large-scale assemblies not possible for SARS-CoV, potentially mediating unique immune suppression and evasion activities.

6.
J Virol ; 93(24)2019 12 15.
Article in English | MEDLINE | ID: mdl-31578291

ABSTRACT

The accessory protein Nef of human immunodeficiency virus (HIV) is a primary determinant of viral pathogenesis. Nef is abundantly expressed during infection and reroutes a variety of cell surface proteins to disrupt host immunity and promote the viral replication cycle. Nef counteracts host defenses by sequestering and/or degrading its targets via the endocytic and secretory pathways. Nef does this by physically engaging a number of host trafficking proteins. Substantial progress has been achieved in identifying the targets of Nef, and a structural and mechanistic understanding of Nef's ability to command the protein trafficking machinery has recently started to coalesce. Comparative analysis of HIV and simian immunodeficiency virus (SIV) Nef proteins in the context of recent structural advances sheds further light on both viral evolution and the mechanisms whereby trafficking is hijacked. This review describes how advances in cell and structural biology are uncovering in growing detail how Nef subverts the host immune system, facilitates virus release, and enhances viral infectivity.


Subject(s)
HIV Infections/immunology , HIV Infections/metabolism , HIV-1/metabolism , Protein Transport/physiology , nef Gene Products, Human Immunodeficiency Virus/metabolism , Clathrin-Coated Vesicles , HIV Infections/virology , HIV-1/immunology , Host-Pathogen Interactions , Humans , Membrane Proteins/metabolism , Models, Molecular , Protein Conformation , Simian Immunodeficiency Virus/immunology , Viral Regulatory and Accessory Proteins/chemistry , nef Gene Products, Human Immunodeficiency Virus/chemistry
7.
Cell Host Microbe ; 26(3): 359-368.e8, 2019 Sep 11.
Article in English | MEDLINE | ID: mdl-31447307

ABSTRACT

Tetherin is a host defense factor that physically prevents virion release from the plasma membrane. The Nef accessory protein of simian immunodeficiency virus (SIV) engages the clathrin adaptor AP-2 to downregulate tetherin via its DIWK motif. As human tetherin lacks DIWK, antagonism of tetherin by Nef is a barrier to simian-human transmission of non-human primate lentiviruses. To determine the molecular basis for tetherin counteraction, we reconstituted the AP-2 complex with a simian tetherin and SIV Nef and determined its structure by cryoelectron microscopy (cryo-EM). Nef refolds the first α-helix of the ß2 subunit of AP-2 to a ß hairpin, creating a binding site for the DIWK sequence. The tetherin binding site in Nef is distinct from those of most other Nef substrates, including MHC class I, CD3, and CD4 but overlaps with the site for the restriction factor SERINC5. This structure explains the dependence of SIVs on tetherin DIWK and consequent barrier to human transmission.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Bone Marrow Stromal Antigen 2/chemistry , Bone Marrow Stromal Antigen 2/pharmacology , Lentivirus Infections/prevention & control , Lentivirus Infections/transmission , Zoonoses/virology , Adaptor Protein Complex 2/chemistry , Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex beta Subunits/chemistry , Animals , Binding Sites , CD3 Complex/metabolism , CD4 Antigens/metabolism , Cell Membrane/drug effects , Cryoelectron Microscopy , Down-Regulation , Gene Products, nef/chemistry , Gene Products, nef/metabolism , HEK293 Cells , Histocompatibility Antigens Class I/metabolism , Humans , Lentivirus Infections/virology , Membrane Proteins/metabolism , Models, Molecular , Primary Cell Culture , Protein Conformation , Protein Conformation, alpha-Helical , Protein Folding , Protein Interaction Domains and Motifs , Sequence Alignment , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/metabolism , Virion/drug effects
8.
Cell ; 174(3): 659-671.e14, 2018 07 26.
Article in English | MEDLINE | ID: mdl-30053425

ABSTRACT

The HIV accessory protein Nef counteracts immune defenses by subverting coated vesicle pathways. The 3.7 Å cryo-EM structure of a closed trimer of the clathrin adaptor AP-1, the small GTPase Arf1, HIV-1 Nef, and the cytosolic tail of the restriction factor tetherin suggested a mechanism for inactivating tetherin by Golgi retention. The 4.3 Å structure of a mutant Nef-induced dimer of AP-1 showed how the closed trimer is regulated by the dileucine loop of Nef. HDX-MS and mutational analysis were used to show how cargo dynamics leads to alternative Arf1 trimerization, directing Nef targets to be either retained at the trans-Golgi or sorted to lysosomes. Phosphorylation of the NL4-3 M-Nef was shown to regulate AP-1 trimerization, explaining how O-Nefs lacking this phosphosite counteract tetherin but most M-Nefs do not. These observations show how the higher-order organization of a vesicular coat can be allosterically modulated to direct cargoes to distinct fates.


Subject(s)
Transcription Factor AP-1/ultrastructure , nef Gene Products, Human Immunodeficiency Virus/metabolism , nef Gene Products, Human Immunodeficiency Virus/ultrastructure , ADP-Ribosylation Factor 1/metabolism , ADP-Ribosylation Factor 1/ultrastructure , Adaptor Proteins, Vesicular Transport , Bone Marrow Stromal Antigen 2/metabolism , Bone Marrow Stromal Antigen 2/ultrastructure , Clathrin , Golgi Apparatus , HEK293 Cells , HIV-1 , Humans , Protein Transport/physiology , Transcription Factor AP-1/metabolism , Transcription Factor AP-1/physiology , nef Gene Products, Human Immunodeficiency Virus/physiology
10.
Nat Microbiol ; 1(11): 16155, 2016 Sep 05.
Article in English | MEDLINE | ID: mdl-27595425

ABSTRACT

No vaccine exists against group A Streptococcus (GAS), a leading cause of worldwide morbidity and mortality. A severe hurdle is the hypervariability of its major antigen, the M protein, with >200 different M types known. Neutralizing antibodies typically recognize M protein hypervariable regions (HVRs) and confer narrow protection. In stark contrast, human C4b-binding protein (C4BP), which is recruited to the GAS surface to block phagocytic killing, interacts with a remarkably large number of M protein HVRs (apparently ∼90%). Such broad recognition is rare, and we discovered a unique mechanism for this through the structure determination of four sequence-diverse M proteins in complexes with C4BP. The structures revealed a uniform and tolerant 'reading head' in C4BP, which detected conserved sequence patterns hidden within hypervariability. Our results open up possibilities for rational therapies that target the M-C4BP interaction, and also inform a path towards vaccine design.


Subject(s)
Antigens, Bacterial/chemistry , Bacterial Outer Membrane Proteins/chemistry , Carrier Proteins/chemistry , Complement C4b-Binding Protein/chemistry , Amino Acid Sequence , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Binding Sites , Carrier Proteins/metabolism , Complement C4b-Binding Protein/metabolism , Complement Inactivator Proteins , Conserved Sequence , Host-Pathogen Interactions , Humans
11.
Proc Natl Acad Sci U S A ; 113(34): 9515-20, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27512043

ABSTRACT

The sequences of M proteins, the major surface-associated virulence factors of the widespread bacterial pathogen group A Streptococcus, are antigenically variable but have in common a strong propensity to form coiled coils. Paradoxically, these sequences are also replete with coiled-coil destabilizing residues. These features are evident in the irregular coiled-coil structure and thermal instability of M proteins. We present an explanation for this paradox through studies of the B repeats of the medically important M1 protein. The B repeats are required for interaction of M1 with fibrinogen (Fg) and consequent proinflammatory activation. The B repeats sample multiple conformations, including intrinsically disordered, dissociated, as well as two alternate coiled-coil conformations: a Fg-nonbinding register 1 and a Fg-binding register 2. Stabilization of M1 in the Fg-nonbinding register 1 resulted in attenuation of Fg binding as expected, but counterintuitively, so did stabilization in the Fg-binding register 2. Strikingly, these register-stabilized M1 proteins gained the ability to bind Fg when they were destabilized by a chaotrope. These results indicate that M1 stability is antithetical to Fg interaction and that M1 conformational dynamics, as specified by destabilizing residues, are essential for interaction. A "capture-and-collapse" model of association accounts for these observations, in which M1 captures Fg through a dynamic conformation and then collapses into a register 2-coiled coil as a result of stabilization provided by binding energy. Our results support the general conclusion that destabilizing residues are evolutionarily conserved in M proteins to enable functional interactions necessary for pathogenesis.


Subject(s)
Amino Acids/chemistry , Antigens, Bacterial/chemistry , Bacterial Outer Membrane Proteins/chemistry , Carrier Proteins/chemistry , Fibrinogen/chemistry , Streptococcus pyogenes/chemistry , Amino Acid Sequence , Amino Acids/metabolism , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Binding Sites , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Fibrinogen/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Mutation , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thermodynamics
12.
J Biol Chem ; 289(46): 32303-32315, 2014 Nov 14.
Article in English | MEDLINE | ID: mdl-25266727

ABSTRACT

A recent analysis of group A Streptococcus (GAS) invasive infections in Australia has shown a predominance of M4 GAS, a serotype recently reported to lack the antiphagocytic hyaluronic acid (HA) capsule. Here, we use molecular genetics and bioinformatics techniques to characterize 17 clinical M4 isolates associated with invasive disease in children during this recent epidemiology. All M4 isolates lacked HA capsule, and whole genome sequence analysis of two isolates revealed the complete absence of the hasABC capsule biosynthesis operon. Conversely, M4 isolates possess a functional HA-degrading hyaluronate lyase (HylA) enzyme that is rendered nonfunctional in other GAS through a point mutation. Transformation with a plasmid expressing hasABC restored partial encapsulation in wild-type (WT) M4 GAS, and full encapsulation in an isogenic M4 mutant lacking HylA. However, partial encapsulation reduced binding to human complement regulatory protein C4BP, did not enhance survival in whole human blood, and did not increase virulence of WT M4 GAS in a mouse model of systemic infection. Bioinformatics analysis found no hasABC homologs in closely related species, suggesting that this operon was a recent acquisition. These data showcase a mutually exclusive interaction of HA capsule and active HylA among strains of this leading human pathogen.


Subject(s)
Hyaluronic Acid/metabolism , Hyaluronoglucosaminidase/metabolism , Streptococcal Infections/microbiology , Streptococcus pyogenes/enzymology , Animals , Bacterial Proteins/metabolism , Base Sequence , Cell Membrane/microbiology , Computational Biology , Exotoxins/metabolism , Female , Genetic Complementation Test , Histidine Kinase , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Molecular Sequence Data , Neutrophils/microbiology , Point Mutation , Polysaccharide-Lyases/metabolism , Polysaccharides/metabolism , Recombinant Proteins/metabolism , Repressor Proteins/metabolism , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...