Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Front Mol Biosci ; 10: 1275393, 2023.
Article in English | MEDLINE | ID: mdl-37681016
2.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Article in English | MEDLINE | ID: mdl-33479176

ABSTRACT

The reversibility of ubiquitination by the action of deubiquitinating enzymes (DUBs) serves as an important regulatory layer within the ubiquitin system. Approximately 100 DUBs are encoded by the human genome, and many have been implicated with pathologies, including neurodegeneration and cancer. Non-lysine ubiquitination is chemically distinct, and its physiological importance is emerging. Here, we couple chemically and chemoenzymatically synthesized ubiquitinated lysine and threonine model substrates to a mass spectrometry-based DUB assay. Using this platform, we profile two-thirds of known catalytically active DUBs for threonine esterase and lysine isopeptidase activity and find that most DUBs demonstrate dual selectivity. However, with two anomalous exceptions, the ovarian tumor domain DUB class demonstrates specific (iso)peptidase activity. Strikingly, we find the Machado-Joseph disease (MJD) class to be unappreciated non-lysine DUBs with highly specific ubiquitin esterase activity rivaling the efficiency of the most active isopeptidases. Esterase activity is dependent on the canonical catalytic triad, but proximal hydrophobic residues appear to be general determinants of non-lysine activity. Our findings also suggest that ubiquitin esters have appreciable cellular stability and that non-lysine ubiquitination is an integral component of the ubiquitin system. Its regulatory sophistication is likely to rival that of canonical ubiquitination.


Subject(s)
Deubiquitinating Enzymes/genetics , Esterases/genetics , Machado-Joseph Disease/genetics , Ubiquitin/genetics , Amino Acids/genetics , Deubiquitinating Enzymes/isolation & purification , Humans , Lysine/genetics , Machado-Joseph Disease/enzymology , Machado-Joseph Disease/pathology , Mass Spectrometry , Protein Processing, Post-Translational/genetics , Ubiquitination/genetics
3.
Nat Chem Biol ; 16(11): 1227-1236, 2020 11.
Article in English | MEDLINE | ID: mdl-32747811

ABSTRACT

MYCBP2 is a ubiquitin (Ub) E3 ligase (E3) that is essential for neurodevelopment and regulates axon maintenance. MYCBP2 transfers Ub to nonlysine substrates via a newly discovered RING-Cys-Relay (RCR) mechanism, where Ub is relayed from an upstream cysteine to a downstream substrate esterification site. The molecular bases for E2-E3 Ub transfer and Ub relay are unknown. Whether these activities are linked to the neural phenotypes is also unclear. We describe the crystal structure of a covalently trapped E2~Ub:MYCBP2 transfer intermediate revealing key structural rearrangements upon E2-E3 Ub transfer and Ub relay. Our data suggest that transfer to the dynamic upstream cysteine, whilst mitigating lysine activity, requires a closed-like E2~Ub conjugate with tempered reactivity, and Ub relay is facilitated by a helix-coil transition. Furthermore, neurodevelopmental defects and delayed injury-induced degeneration in RCR-defective knock-in mice suggest its requirement, and that of substrate esterification activity, for normal neural development and programmed axon degeneration.


Subject(s)
Axons/metabolism , Cysteine/metabolism , RING Finger Domains , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Binding Sites , Female , Gene Knock-In Techniques , Humans , Lysine/metabolism , Mice , Mice, Inbred C57BL/embryology , Mice, Transgenic , Models, Molecular , Molecular Conformation , Protein Binding , Protein Conformation , Signal Transduction , Structure-Activity Relationship , Ubiquitination
4.
Cell Chem Biol ; 27(1): 74-82.e6, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31859248

ABSTRACT

Activity-based protein profiling is an invaluable technique for studying enzyme biology and facilitating the development of therapeutics. Ubiquitin E3 ligases (E3s) are one of the largest enzyme families and regulate a host of (patho)physiological processes. The largest subtype are the RING E3s of which there are >600 members. RING E3s have adaptor-like activity that can be subject to diverse regulatory mechanisms and have become attractive drug targets. Activity-based probes (ABPs) for measuring RING E3 activity do not exist. Here we re-engineer ubiquitin-charged E2 conjugating enzymes to produce photocrosslinking ABPs. We demonstrate activity-dependent profiling of two divergent cancer-associated RING E3s, RNF4 and c-Cbl, in response to their native activation signals. We also demonstrate profiling of endogenous RING E3 ligase activation in response to epidermal growth factor (EGF) stimulation. These photocrosslinking ABPs should advance E3 ligase research and the development of selective modulators against this important class of enzymes.


Subject(s)
Benzophenones/chemistry , Cross-Linking Reagents/chemistry , Phenylalanine/analogs & derivatives , Ubiquitin-Protein Ligases/metabolism , Benzophenones/metabolism , Cross-Linking Reagents/metabolism , Humans , Models, Molecular , Molecular Conformation , Phenylalanine/chemistry , Phenylalanine/metabolism , Photochemical Processes , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/chemistry
5.
Cell Rep ; 29(12): 3983-3996.e4, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31851928

ABSTRACT

HIV-1 hijacks host proteins to promote infection. Here we show that HIV is also dependent upon the host metabolite inositol hexakisphosphate (IP6) for viral production and primary cell replication. HIV-1 recruits IP6 into virions using two lysine rings in its immature hexamers. Mutation of either ring inhibits IP6 packaging and reduces viral production. Loss of IP6 also results in virions with highly unstable capsids, leading to a profound loss of reverse transcription and cell infection. Replacement of one ring with a hydrophobic isoleucine core restores viral production, but IP6 incorporation and infection remain impaired, consistent with an independent role for IP6 in stable capsid assembly. Genetic knockout of biosynthetic kinases IPMK and IPPK reveals that cellular IP6 availability limits the production of diverse lentiviruses, but in the absence of IP6, HIV-1 packages IP5 without loss of infectivity. Together, these data suggest that IP6 is a critical cofactor for HIV-1 replication.


Subject(s)
Capsid/metabolism , HIV Infections/virology , HIV-1/physiology , Host-Pathogen Interactions , Phytic Acid/metabolism , Virus Assembly , Virus Replication , Capsid/chemistry , HIV Infections/metabolism , HIV Infections/pathology , HeLa Cells , Humans , Protein Conformation
6.
Cell Host Microbe ; 24(6): 761-775.e6, 2018 12 12.
Article in English | MEDLINE | ID: mdl-30503508

ABSTRACT

TRIM5 is a RING domain E3 ubiquitin ligase with potent antiretroviral function. TRIM5 assembles into a hexagonal lattice on retroviral capsids, causing envelopment of the infectious core. Concomitantly, TRIM5 initiates innate immune signaling and orchestrates disassembly of the viral particle, yet how these antiviral responses are regulated by capsid recognition is unclear. We show that hexagonal assembly triggers N-terminal polyubiquitination of TRIM5 that collectively drives antiviral responses. In uninfected cells, N-terminal monoubiquitination triggers non-productive TRIM5 turnover. Upon TRIM5 assembly on virus, a trivalent RING arrangement allows elongation of N-terminally anchored K63-linked ubiquitin chains (N-K63-Ub). N-K63-Ub drives TRIM5 innate immune stimulation and proteasomal degradation. Inducing ubiquitination before TRIM5 assembly triggers premature degradation and ablates antiviral restriction. Conversely, driving N-K63 ubiquitination after TRIM5 assembly enhances innate immune signaling. Thus, the hexagonal geometry of TRIM5's antiviral lattice converts a capsid-binding protein into a multifunctional antiviral platform.


Subject(s)
Carrier Proteins/metabolism , Immunity, Innate/immunology , Retroviridae Infections/immunology , Ubiquitin-Protein Ligases/metabolism , Animals , Antiviral Restriction Factors , Capsid/chemistry , Capsid/metabolism , Carrier Proteins/genetics , HEK293 Cells , Humans , Leukemia Virus, Murine/enzymology , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/immunology , Mice , Mice, Inbred C57BL , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/genetics , Polysaccharides, Bacterial/metabolism , Retroviridae Infections/metabolism , Retroviridae Infections/virology , THP-1 Cells , Tripartite Motif Proteins , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics
7.
Sci Signal ; 11(526)2018 04 17.
Article in English | MEDLINE | ID: mdl-29666306

ABSTRACT

A major challenge in cancer genomics is identifying "driver" mutations from the many neutral "passenger" mutations within a given tumor. To identify driver mutations that would otherwise be lost within mutational noise, we filtered genomic data by motifs that are critical for kinase activity. In the first step of our screen, we used data from the Cancer Cell Line Encyclopedia and The Cancer Genome Atlas to identify kinases with truncation mutations occurring within or before the kinase domain. The top 30 tumor-suppressing kinases were aligned, and hotspots for loss-of-function (LOF) mutations were identified on the basis of amino acid conservation and mutational frequency. The functional consequences of new LOF mutations were biochemically validated, and the top 15 hotspot LOF residues were used in a pan-cancer analysis to define the tumor-suppressing kinome. A ranked list revealed MAP2K7, an essential mediator of the c-Jun N-terminal kinase (JNK) pathway, as a candidate tumor suppressor in gastric cancer, despite its mutational frequency falling within the mutational noise for this cancer type. The majority of mutations in MAP2K7 abolished its catalytic activity, and reactivation of the JNK pathway in gastric cancer cells harboring LOF mutations in MAP2K7 or the downstream kinase JNK suppressed clonogenicity and growth in soft agar, demonstrating the functional relevance of inactivating the JNK pathway in gastric cancer. Together, our data highlight a broadly applicable strategy to identify functional cancer driver mutations and define the JNK pathway as tumor-suppressive in gastric cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Genomics/methods , Loss of Function Mutation , MAP Kinase Kinase 7/genetics , MAP Kinase Signaling System/genetics , Stomach Neoplasms/genetics , Amino Acid Sequence , Cell Line, Tumor , Genes, Tumor Suppressor , Humans , MAP Kinase Kinase 7/chemistry , MAP Kinase Kinase 7/metabolism , Molecular Dynamics Simulation , Sequence Homology, Amino Acid , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology
8.
Elife ; 72018 04 18.
Article in English | MEDLINE | ID: mdl-29667579

ABSTRACT

Cell surface Fc receptors activate inflammation and are tightly controlled to prevent autoimmunity. Antibodies also simulate potent immune signalling from inside the cell via the cytosolic antibody receptor TRIM21, but how this is regulated is unknown. Here we show that TRIM21 signalling is constitutively repressed by its B-Box domain and activated by phosphorylation. The B-Box occupies an E2 binding site on the catalytic RING domain by mimicking E2-E3 interactions, inhibiting TRIM21 ubiquitination and preventing immune activation. TRIM21 is derepressed by IKKß and TBK1 phosphorylation of an LxxIS motif in the RING domain, at the interface with the B-Box. Incorporation of phosphoserine or a phosphomimetic within this motif relieves B-Box inhibition, promoting E2 binding, RING catalysis, NF-κB activation and cytokine transcription upon infection with DNA or RNA viruses. These data explain how intracellular antibody signalling is regulated and reveal that the B-Box is a critical regulator of RING E3 ligase activity.


Subject(s)
Gene Expression Regulation , Protein Processing, Post-Translational , Receptors, Fc/metabolism , Ribonucleoproteins/metabolism , Signal Transduction , Animals , Cell Line , Humans , I-kappa B Kinase/metabolism , Mice , Phosphorylation , Protein Serine-Threonine Kinases/metabolism
9.
J Virol ; 90(10): 4856-4859, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26937031

ABSTRACT

TRIM21 is a high-affinity antibody receptor uniquely expressed in the cytosol of mammalian cells. Here we summarize its role in extending antibody protection into the intracellular environment and allowing nonprofessional cells to benefit from adaptive immunity. We highlight recent work that has shed light on how TRIM21 acts as both an immune sensor and effector. We also review how TRIM21 synergizes with other innate immune receptors to promote an integrated antiviral response.


Subject(s)
Adaptive Immunity , Antibodies, Neutralizing/immunology , Immunity, Innate , Receptors, IgG/immunology , Ribonucleoproteins/immunology , Ribonucleoproteins/metabolism , Virus Diseases/immunology , Animals , Antibodies, Neutralizing/biosynthesis , Antigen-Antibody Complex/immunology , Cytosol/chemistry , Cytosol/immunology , Humans , Mice , Receptors, IgG/metabolism , Ubiquitin/metabolism
10.
Proc Natl Acad Sci U S A ; 112(32): 10014-9, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26150489

ABSTRACT

Tripartite motif (TRIM) 21 is a cytosolic antibody receptor that neutralizes antibody-coated viruses that penetrate the cell and simultaneously activates innate immunity. Here we show that the conjugation of TRIM21 with K63-linked ubiquitin (Ub-(63)Ub) catalyzed by the sequential activity of nonredundant E2 Ub enzymes is required for its dual antiviral functions. TRIM21 is first labeled with monoubiquitin (monoUb) by the E2 Ube2W. The monoUb is a substrate for the heterodimeric E2 Ube2N/Ube2V2, resulting in TRIM21-anchored Ub-(63)Ub. Depletion of either E2 abolishes Ub-(63)Ub and Ub-(48)Ub conjugation of TRIM21, NF-κB signaling, and virus neutralization. The formation of TRIM21-Ub-(63)Ub precedes proteasome recruitment, and we identify an essential role for the 19S-resident and degradation-coupled deubiquitinase Poh1 in TRIM21 neutralization, signaling, and cytokine induction. This study elucidates a complex mechanism of step-wise ubiquitination and deubiquitination activities that allows contemporaneous innate immune signaling and neutralization by TRIM21.


Subject(s)
Ribonucleoproteins/metabolism , Ubiquitination , Animals , Cell Line , Cytokines/genetics , Humans , Lysine/metabolism , Mice , Models, Biological , NF-kappa B/metabolism , Neutralization Tests , Polyubiquitin/metabolism , Proteasome Endopeptidase Complex/metabolism , Signal Transduction , Substrate Specificity , Trans-Activators/metabolism , Transcription, Genetic , Ubiquitin-Conjugating Enzymes/metabolism
11.
EMBO J ; 34(15): 2078-95, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26101372

ABSTRACT

TRIM5α is an antiviral, cytoplasmic, E3 ubiquitin (Ub) ligase that assembles on incoming retroviral capsids and induces their premature dissociation. It inhibits reverse transcription of the viral genome and can also synthesize unanchored polyubiquitin (polyUb) chains to stimulate innate immune responses. Here, we show that TRIM5α employs the E2 Ub-conjugating enzyme Ube2W to anchor the Lys63-linked polyUb chains in a process of TRIM5α auto-ubiquitination. Chain anchoring is initiated, in cells and in vitro, through Ube2W-catalyzed monoubiquitination of TRIM5α. This modification serves as a substrate for the elongation of anchored Lys63-linked polyUb chains, catalyzed by the heterodimeric E2 enzyme Ube2N/Ube2V2. Ube2W targets multiple TRIM5α internal lysines with Ub especially lysines 45 and 50, rather than modifying the N-terminal amino group, which is instead αN-acetylated in cells. E2 depletion or Ub mutation inhibits TRIM5α ubiquitination in cells and restores restricted viral reverse transcription, but not infection. Our data indicate that the stepwise formation of anchored Lys63-linked polyUb is a critical early step in the TRIM5α restriction mechanism and identify the E2 Ub-conjugating cofactors involved.


Subject(s)
Carrier Proteins/metabolism , Models, Biological , Reverse Transcription/physiology , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin/metabolism , Antiviral Restriction Factors , HEK293 Cells , HeLa Cells , Humans , Mutagenesis, Site-Directed , RNA Interference , RNA, Small Interfering/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases
12.
PLoS Pathog ; 10(10): e1004459, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25356722

ABSTRACT

The HIV-1 capsid is involved in all infectious steps from reverse transcription to integration site selection, and is the target of multiple host cell and pharmacologic ligands. However, structural studies have been limited to capsid monomers (CA), and the mechanistic basis for how these ligands influence infection is not well understood. Here we show that a multi-subunit interface formed exclusively within CA hexamers mediates binding to linear epitopes within cellular cofactors NUP153 and CPSF6, and is competed for by the antiretroviral compounds PF74 and BI-2. Each ligand is anchored via a shared phenylalanine-glycine (FG) motif to a pocket within the N-terminal domain of one monomer, and all but BI-2 also make essential interactions across the N-terminal domain: C-terminal domain (NTD:CTD) interface to a second monomer. Dissociation of hexamer into CA monomers prevents high affinity interaction with CPSF6 and PF74, and abolishes binding to NUP153. The second interface is conformationally dynamic, but binding of NUP153 or CPSF6 peptides is accommodated by only one conformation. NUP153 and CPSF6 have overlapping binding sites, but each makes unique CA interactions that, when mutated selectively, perturb cofactor dependency. These results reveal that multiple ligands share an overlapping interface in HIV-1 capsid that is lost upon viral disassembly.


Subject(s)
Capsid Proteins/metabolism , Capsid/chemistry , HIV Infections/virology , HIV-1/chemistry , Nuclear Pore Complex Proteins/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Anti-HIV Agents/pharmacology , Binding Sites , Capsid/metabolism , Capsid Proteins/genetics , HIV Infections/drug therapy , HIV-1/genetics , HIV-1/metabolism , Humans , Indoles/pharmacology , Ligands , Models, Molecular , Models, Structural , Mutation , Nuclear Pore Complex Proteins/genetics , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Polycyclic Compounds/pharmacology , Polymerization , Protein Binding , Protein Structure, Tertiary , Reverse Transcription/drug effects , Virion , mRNA Cleavage and Polyadenylation Factors/genetics
13.
Arthritis Rheumatol ; 66(11): 2976-88, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25132672

ABSTRACT

OBJECTIVE: HLA-B27 forms misfolded heavy chain dimers, which may predispose individuals to inflammatory arthritis by inducing endoplasmic reticulum (ER) stress and the unfolded protein response (UPR). This study was undertaken to define the role of the UPR-induced ER-associated degradation (ERAD) pathway in the disposal of HLA-B27 dimeric conformers. METHODS: HeLa cell lines expressing only 2 copies of a carboxy-terminally Sv5-tagged HLA-B27 were generated. The ER stress-induced protein ER degradation-enhancing α-mannosidase-like protein 1 (EDEM1) was overexpressed by transfection, and dimer levels were monitored by immunoblotting. EDEM1, the UPR-associated transcription factor X-box binding protein 1 (XBP-1), the E3 ubiquitin ligase hydroxymethylglutaryl-coenzyme A reductase degradation 1 (HRD1), and the degradation-associated proteins derlin 1 and derlin 2 were inhibited using either short hairpin RNA or dominant-negative mutants. The UPR-associated ERAD of HLA-B27 was confirmed using ER stress-inducing pharamacologic agents in kinetic and pulse chase assays. RESULTS: We demonstrated that UPR-induced machinery can target HLA-B27 dimers and that dimer formation can be controlled by alterations to expression levels of components of the UPR-induced ERAD pathway. HLA-B27 dimers and misfolded major histocompatibility complex class I monomeric molecules bound to EDEM1 were detected, and overexpression of EDEM1 led to inhibition of HLA-B27 dimer formation. EDEM1 inhibition resulted in up-regulation of HLA-B27 dimers, while UPR-induced ERAD of dimers was prevented in the absence of EDEM1. HLA-B27 dimer formation was also enhanced in the absence of XBP-1, HRD1, and derlins 1 and 2. CONCLUSION: The present findings indicate that the UPR ERAD pathway can dispose of HLA-B27 dimers, thus presenting a potential novel therapeutic target for modulation of HLA-B27-associated inflammatory disease.


Subject(s)
Endoplasmic Reticulum-Associated Degradation/physiology , Endoplasmic Reticulum/physiology , HLA-B27 Antigen/physiology , Membrane Proteins/physiology , Protein Folding , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/physiology , HeLa Cells , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/drug effects , RNA, Small Interfering/pharmacology , Regulatory Factor X Transcription Factors , Signal Transduction/physiology , Transcription Factors/antagonists & inhibitors , Transcription Factors/drug effects , Transcription Factors/physiology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/drug effects , Ubiquitin-Protein Ligases/physiology , X-Box Binding Protein 1
14.
Nature ; 503(7476): 402-405, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24196705

ABSTRACT

Human immunodeficiency virus (HIV)-1 is able to replicate in primary human macrophages without stimulating innate immunity despite reverse transcription of genomic RNA into double-stranded DNA, an activity that might be expected to trigger innate pattern recognition receptors. We reasoned that if correctly orchestrated HIV-1 uncoating and nuclear entry is important for evasion of innate sensors then manipulation of specific interactions between HIV-1 capsid and host factors that putatively regulate these processes should trigger pattern recognition receptors and stimulate type 1 interferon (IFN) secretion. Here we show that HIV-1 capsid mutants N74D and P90A, which are impaired for interaction with cofactors cleavage and polyadenylation specificity factor subunit 6 (CPSF6) and cyclophilins (Nup358 and CypA), respectively, cannot replicate in primary human monocyte-derived macrophages because they trigger innate sensors leading to nuclear translocation of NF-κB and IRF3, the production of soluble type 1 IFN and induction of an antiviral state. Depletion of CPSF6 with short hairpin RNA expression allows wild-type virus to trigger innate sensors and IFN production. In each case, suppressed replication is rescued by IFN-receptor blockade, demonstrating a role for IFN in restriction. IFN production is dependent on viral reverse transcription but not integration, indicating that a viral reverse transcription product comprises the HIV-1 pathogen-associated molecular pattern. Finally, we show that we can pharmacologically induce wild-type HIV-1 infection to stimulate IFN secretion and an antiviral state using a non-immunosuppressive cyclosporine analogue. We conclude that HIV-1 has evolved to use CPSF6 and cyclophilins to cloak its replication, allowing evasion of innate immune sensors and induction of a cell-autonomous innate immune response in primary human macrophages.


Subject(s)
HIV-1/immunology , Immune Evasion , Immunity, Innate/immunology , Macrophages/immunology , Macrophages/virology , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cyclophilins/metabolism , Cyclosporine/metabolism , HIV Infections/immunology , HIV Infections/metabolism , HIV Infections/pathology , HIV Infections/virology , HIV-1/metabolism , Humans , Interferon Regulatory Factor-3/metabolism , Interferon Type I/immunology , Interferon Type I/metabolism , Macrophages/cytology , Macrophages/pathology , Molecular Chaperones/metabolism , Monocytes/cytology , NF-kappa B/metabolism , Nuclear Pore Complex Proteins/metabolism , Receptors, Pattern Recognition , Virus Internalization , Virus Replication/immunology , mRNA Cleavage and Polyadenylation Factors/deficiency , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism
15.
Curr Top Microbiol Immunol ; 371: 29-66, 2013.
Article in English | MEDLINE | ID: mdl-23686231

ABSTRACT

The TRIM protein family is emerging as a central component of mammalian antiviral innate immunity. Beginning with the identification of TRIM5α as a mammalian post-entry restriction factor against retroviruses, to the repeated observation that many TRIMs ubiquitinate and regulate signaling pathways, the past decade has witnessed an intense research effort to understand how TRIM proteins influence immunity. The list of viral families targeted directly or indirectly by TRIM proteins has grown to include adenoviruses, hepadnaviruses, picornaviruses, flaviviruses, orthomyxoviruses, paramyxoviruses, herpesviruses, rhabdoviruses and arenaviruses. We have come to appreciate how, through intense bouts of positive selection, some TRIM genes have been honed into species-specific restriction factors. Similarly, in the case of TRIMCyp, we are beginning to understand how viruses too have mutated to evade restriction, suggesting that TRIM and viruses have coevolved for millions of years of primate evolution. Recently, TRIM5α returned to the limelight when it was shown to trigger the expression of antiviral genes upon recognition of an incoming virus, a paradigm shift that demonstrated that restriction factors make excellent pathogen sensors. However, it remains unclear how many of ~100 human TRIM genes are antiviral, despite the expression of many of these genes being upregulated by interferon and upon viral infection. TRIM proteins do not conform to one type of antiviral mechanism, reflecting the diversity of viruses they target. Moreover, the cofactors of restriction remain largely enigmatic. The control of retroviral replication remains an important medical subject and provides a useful backdrop for reviewing how TRIM proteins act to repress viral replication.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Immunity, Innate/immunology , Membrane Proteins/immunology , Retroviridae/physiology , Virus Replication/immunology , Animals , Humans
16.
PLoS Pathog ; 8(8): e1002896, 2012.
Article in English | MEDLINE | ID: mdl-22956906

ABSTRACT

The HIV-1 genome enters cells inside a shell comprised of capsid (CA) protein. Variation in CA sequence alters HIV-1 infectivity and escape from host restriction factors. However, apart from the Cyclophilin A-binding loop, CA has no known interfaces with which to interact with cellular cofactors. Here we describe a novel protein-protein interface in the N-terminal domain of HIV-1 CA, determined by X-ray crystallography, which mediates both viral restriction and host cofactor dependence. The interface is highly conserved across lentiviruses and is accessible in the context of a hexameric lattice. Mutation of the interface prevents binding to and restriction by CPSF6-358, a truncated cytosolic form of the RNA processing factor, cleavage and polyadenylation specific factor 6 (CPSF6). Furthermore, mutations that prevent CPSF6 binding also relieve dependence on nuclear entry cofactors TNPO3 and RanBP2. These results suggest that the HIV-1 capsid mediates direct host cofactor interactions to facilitate viral infection.


Subject(s)
Capsid Proteins/metabolism , HIV Infections/virology , HIV-1/physiology , Virus Replication , mRNA Cleavage and Polyadenylation Factors/metabolism , Amino Acid Sequence , Antiviral Agents/metabolism , Capsid Proteins/chemistry , Capsid Proteins/genetics , Cell Line, Tumor , Conserved Sequence , Crystallography, X-Ray , HIV-1/genetics , Humans , Indoles/metabolism , Models, Molecular , Molecular Chaperones/metabolism , Molecular Sequence Data , Mutation , Nuclear Pore Complex Proteins/metabolism , Phenylalanine/analogs & derivatives , Phenylalanine/metabolism , Protein Binding , Sequence Alignment , Virus Internalization , beta Karyopherins/metabolism , mRNA Cleavage and Polyadenylation Factors/genetics
17.
PLoS Pathog ; 7(12): e1002439, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22174692

ABSTRACT

Lentiviruses such as HIV-1 traverse nuclear pore complexes (NPC) and infect terminally differentiated non-dividing cells, but how they do this is unclear. The cytoplasmic NPC protein Nup358/RanBP2 was identified as an HIV-1 co-factor in previous studies. Here we report that HIV-1 capsid (CA) binds directly to the cyclophilin domain of Nup358/RanBP2. Fusion of the Nup358/RanBP2 cyclophilin (Cyp) domain to the tripartite motif of TRIM5 created a novel inhibitor of HIV-1 replication, consistent with an interaction in vivo. In contrast to CypA binding to HIV-1 CA, Nup358 binding is insensitive to inhibition with cyclosporine, allowing contributions from CypA and Nup358 to be distinguished. Inhibition of CypA reduced dependence on Nup358 and the nuclear basket protein Nup153, suggesting that CypA regulates the choice of the nuclear import machinery that is engaged by the virus. HIV-1 cyclophilin-binding mutants CA G89V and P90A favored integration in genomic regions with a higher density of transcription units and associated features than wild type virus. Integration preference of wild type virus in the presence of cyclosporine was similarly altered to regions of higher transcription density. In contrast, HIV-1 CA alterations in another patch on the capsid surface that render the virus less sensitive to Nup358 or TRN-SR2 depletion (CA N74D, N57A) resulted in integration in genomic regions sparse in transcription units. Both groups of CA mutants are impaired in replication in HeLa cells and human monocyte derived macrophages. Our findings link HIV-1 engagement of cyclophilins with both integration targeting and replication efficiency and provide insight into the conservation of viral cyclophilin recruitment.


Subject(s)
Capsid Proteins/metabolism , Cell Nucleus/virology , Cyclophilin A/metabolism , HIV Infections/metabolism , HIV-1/physiology , Virus Replication , Active Transport, Cell Nucleus/physiology , Blotting, Western , Cell Line , Cell Nucleus/metabolism , HeLa Cells , Humans , Macrophages/metabolism , Macrophages/virology , Molecular Chaperones/metabolism , Nuclear Pore Complex Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Virus Replication/physiology
18.
J Virol ; 84(23): 12463-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20861252

ABSTRACT

TRIM5α proteins recruit and restrict incoming cytoplasmic retroviruses. Primate TRIM5α sequence diversity underlies species-specific restriction and is likely caused by selective pressure from ancient pathogenic infections. Here we show that TRIM5α from the European brown hare restricts diverse retroviruses. Furthermore, it differs significantly in sequence from TRIM5α from the closely related rabbit, suggesting evolutionary changes in the last 12 million years since these species diverged. We propose that, like primates, lagomorphs have been subject to selective pressure from TRIM5-sensitive viruses, possibly related to the endogenous lentivirus RELIK found in both rabbits and hares.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Genetic Variation , Hares/virology , Retroviridae/metabolism , Amino Acid Sequence , Animals , Base Sequence , Capsid Proteins/metabolism , Cloning, Molecular , DNA Primers/genetics , Evolution, Molecular , Genetic Vectors , Green Fluorescent Proteins , Hares/genetics , Likelihood Functions , Models, Genetic , Molecular Sequence Data , Sequence Alignment , Sequence Analysis, DNA , Species Specificity
19.
J Virol ; 83(4): 2044-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19073742

ABSTRACT

Cyclophilin A (CypA) is an important human immunodeficiency virus type 1 (HIV-1) cofactor in human cells. HIV-1 A92E and G94D capsid escape mutants arise during CypA inhibition and in certain cell lines are dependent on CypA inhibition. Here we show that dependence on CypA inhibition is due to high CypA levels. Restricted HIV-1 is stable, and remarkably, restriction is augmented by arresting cell division. Nuclear entry is not inhibited. We propose that high CypA levels and capsid mutations combine to disturb uncoating, leading to poor infectivity, particularly in arrested cells. Our data suggest a role for CypA in uncoating the core of HIV-1 to facilitate integration.


Subject(s)
Capsid Proteins/genetics , Capsid Proteins/immunology , Cyclophilin A/immunology , HIV-1/physiology , Mutation, Missense , HIV-1/genetics , HeLa Cells , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...