Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Mol Biol ; 436(4): 168409, 2024 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-38128824

RESUMEN

Human immunodeficiency virus type 1 (HIV-1) stimulates innate immune responses upon infection, including cyclic GMP-AMP synthase (cGAS) signaling that results in type I interferon production. HIV-1-induced activation of cGAS requires the host cell factor polyglutamine binding protein 1 (PQBP1), an intrinsically disordered protein that bridges capsid recognition and cGAS recruitment. However, the molecular details of PQBP1 interactions with the HIV-1 capsid and their functional implications remain poorly understood. Here, we show that PQBP1 binds to HIV-1 capsids through charge complementing contacts between acidic residues in the N-terminal region of PQBP1 and an arginine ring in the central channel of the HIV-1 CA hexamer that makes up the viral capsid. These studies reveal the molecular details of PQBP1's primary interaction with the HIV-1 capsid and suggest that additional elements are likely to contribute to stable capsid binding.


Asunto(s)
Cápside , Proteínas de Unión al ADN , VIH-1 , Humanos , Cápside/química , Proteínas de la Cápside/química , Proteínas de Unión al ADN/química , VIH-1/química , Inmunidad Innata , Nucleotidiltransferasas/química , Unión Proteica , Conformación Proteica
2.
Nat Commun ; 14(1): 1237, 2023 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-36871077

RESUMEN

HIV-1 maturation inhibitors (MIs), Bevirimat (BVM) and its analogs interfere with the catalytic cleavage of spacer peptide 1 (SP1) from the capsid protein C-terminal domain (CACTD), by binding to and stabilizing the CACTD-SP1 region. MIs are under development as alternative drugs to augment current antiretroviral therapies. Although promising, their mechanism of action and associated virus resistance pathways remain poorly understood at the molecular, biochemical, and structural levels. We report atomic-resolution magic-angle-spinning NMR structures of microcrystalline assemblies of CACTD-SP1 complexed with BVM and/or the assembly cofactor inositol hexakisphosphate (IP6). Our results reveal a mechanism by which BVM disrupts maturation, tightening the 6-helix bundle pore and quenching the motions of SP1 and the simultaneously bound IP6. In addition, BVM-resistant SP1-A1V and SP1-V7A variants exhibit distinct conformational and binding characteristics. Taken together, our study provides a structural explanation for BVM resistance as well as guidance for the design of new MIs.


Asunto(s)
VIH-1 , Triterpenos , Cápside , Proteínas de la Cápside , Catálisis
3.
Acta Crystallogr F Struct Biol Commun ; 79(Pt 2): 45-50, 2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36748341

RESUMEN

Niemann-Pick C1 protein (NPC1) is a membrane protein that primarily resides in late endosomes and lysosomes, and plays an important role in cholesterol homeostasis in the cell. The second luminal domain of NPC1 (NPC1-C) serves as the intracellular receptor for Ebola and Marburg viruses. Here, the recombinant production of nonglycosylated and glycosylated NPC1-C and a new crystal form of the nonglycosylated protein are reported. The crystals belonged to space group P21 and diffracted to 2.3 Šresolution. The structure is similar to other reported structures of NPC1-C, with differences observed in the protruding loops when compared with NPC1-C in complex with Ebola virus glycoprotein or NPC2.


Asunto(s)
Glicoproteínas de Membrana , Proteína Niemann-Pick C1 , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Proteína Niemann-Pick C1/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Cristalografía por Rayos X , Glicoproteínas/química , Lisosomas/metabolismo
4.
Nat Struct Mol Biol ; 30(3): 383-390, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36759579

RESUMEN

The HIV-1 capsid is a fullerene cone made of quasi-equivalent hexamers and pentamers of the viral CA protein. Typically, quasi-equivalent assembly of viral capsid subunits is controlled by a molecular switch. Here, we identify a Thr-Val-Gly-Gly motif that modulates CA hexamer/pentamer switching by folding into a 310 helix in the pentamer and random coil in the hexamer. Manipulating the coil/helix configuration of the motif allowed us to control pentamer and hexamer formation in a predictable manner, thus proving its function as a molecular switch. Importantly, the switch also remodels the common binding site for host factors that are critical for viral replication and the new ultra-potent HIV-1 inhibitor lenacapavir. This study reveals that a critical assembly element also modulates the post-assembly and viral replication functions of the HIV-1 capsid and provides new insights on capsid function and inhibition.


Asunto(s)
Cápside , VIH-1 , Cápside/química , VIH-1/química , Proteínas de la Cápside/química
5.
Proc Natl Acad Sci U S A ; 119(10): e2117781119, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35238630

RESUMEN

SignificanceThe mature capsids of HIV-1 are transiently stable complexes that self-assemble around the viral genome during maturation, and uncoat to release preintegration complexes that archive a double-stranded DNA copy of the virus in the host cell genome. However, a detailed view of how HIV cores rupture remains lacking. Here, we elucidate the physical properties involved in capsid rupture using a combination of large-scale all-atom molecular dynamics simulations and cryo-electron tomography. We find that intrinsic strain on the capsid forms highly correlated patterns along the capsid surface, along which cracks propagate. Capsid rigidity also increases with high strain. Our findings provide fundamental insight into viral capsid uncoating.


Asunto(s)
Cápside/fisiología , VIH-1/fisiología , Desencapsidación Viral , Cápside/química , Proteínas de la Cápside/química , Línea Celular , Tomografía con Microscopio Electrónico/métodos , Humanos , Simulación de Dinámica Molecular , Conformación Proteica
6.
PLoS Pathog ; 18(2): e1009202, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35130321

RESUMEN

Zinc-finger antiviral protein (ZAP), also known as poly(ADP-ribose) polymerase 13 (PARP13), is an antiviral factor that selectively targets viral RNA for degradation. ZAP is active against both DNA and RNA viruses, including important human pathogens such as hepatitis B virus and type 1 human immunodeficiency virus (HIV-1). ZAP selectively binds CpG dinucleotides through its N-terminal RNA-binding domain, which consists of four zinc fingers. ZAP also contains a central region that consists of a fifth zinc finger and two WWE domains. Through structural and biochemical studies, we found that the fifth zinc finger and tandem WWEs of ZAP combine into a single integrated domain that binds to poly(ADP-ribose) (PAR), a cellular polynucleotide. PAR binding is mediated by the second WWE module of ZAP and likely involves specific recognition of an adenosine diphosphate-containing unit of PAR. Mutation of the PAR binding site in ZAP abrogates the interaction in vitro and diminishes ZAP activity against a CpG-rich HIV-1 reporter virus and murine leukemia virus. In cells, PAR facilitates formation of non-membranous sub-cellular compartments such as DNA repair foci, spindle poles and cytosolic RNA stress granules. Our results suggest that ZAP-mediated viral mRNA degradation is facilitated by PAR, and provides a biophysical rationale for the reported association of ZAP with RNA stress granules.


Asunto(s)
VIH-1/metabolismo , Virus de la Leucemia Murina/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Dedos de Zinc , Animales , Antivirales/farmacología , Cristalografía por Rayos X , Células HEK293 , Células HeLa , Humanos , Ratones , Mutación , Unión Proteica , Conformación Proteica , Dominios Proteicos , Estabilidad del ARN , ARN Viral , Proteínas de Unión al ARN/farmacología
7.
Proteins ; 90(1): 309-313, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34357660

RESUMEN

The Gag proteins of retroviruses play an essential role in virus particle assembly by forming a protein shell or capsid and thus generating the virion compartment. A variety of human proteins have now been identified with structural similarity to one or more of the major Gag domains. These human proteins are thought to have been evolved or "domesticated" from ancient integrations due to retroviral infections or retrotransposons. Here, we report that X-ray crystal structures of stably folded domains of MOAP1 (modulator of apoptosis 1) and PEG10 (paternally expressed gene 10) are highly similar to the C-terminal capsid (CA) domains of cognate Gag proteins. The structures confirm classification of MOAP1 and PEG10 as domesticated Gags, and suggest that these proteins may have preserved some of the key interactions that facilitated assembly of their ancestral Gags into capsids.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Reguladoras de la Apoptosis , Proteínas de Unión al ADN , Productos del Gen gag , Proteínas de Unión al ARN , Retroelementos/genética , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Aminoácidos/genética , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/genética , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Secuencia Conservada/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Productos del Gen gag/química , Productos del Gen gag/genética , Humanos , Modelos Moleculares , Dominios Proteicos/genética , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Retroviridae/genética , Infecciones por Retroviridae
8.
Front Mol Biosci ; 8: 767040, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34957215

RESUMEN

Histidine residues play important structural and functional roles in proteins, such as serving as metal-binding ligands, mediating enzyme catalysis, and modulating proton channel activity. Many of these activities are modulated by the ionization state of the imidazole ring. Here we present a fast MAS NMR approach for the determination of protonation and tautomeric states of His at frequencies of 40-62 kHz. The experiments combine 1H detection with selective magnetization inversion techniques and transferred echo double resonance (TEDOR)-based filters, in 2D heteronuclear correlation experiments. We illustrate this approach using microcrystalline assemblies of HIV-1 CACTD-SP1 protein.

9.
J Virol ; 95(19): e0061521, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34287037

RESUMEN

The transition from an immature to a fully infectious mature retrovirus particle is associated with molecular switches that trigger dramatic conformational changes in the structure of the Gag proteins. A dominant maturation switch that stabilizes the immature capsid (CA) lattice is located downstream of the CA protein in many retroviral Gags. The HIV-1 Gag protein contains a stretch of 5 amino acid residues termed the "clasp motif," important for the organization of the hexameric subunits that provide stability to the overall immature HIV-1 shell. Sequence alignment of the CA C-terminal domains (CTDs) of HIV-1 and Mason-Pfizer monkey virus (M-PMV) highlighted a spacer-like domain in M-PMV that may provide a comparable function. The importance of the sequences spanning the CA-nucleocapsid (NC) cleavage has been demonstrated by mutagenesis, but the specific requirements for the clasp motif in several steps of M-PMV particle assembly and maturation have not been determined in detail. In the present study, we report an examination of the role of the clasp motif in the M-PMV life cycle. We generated a series of M-PMV Gag mutants and assayed for assembly of the recombinant proteins in vitro and for the assembly, maturation, release, genomic RNA packaging, and infectivity of the mutant viruses in vivo. The mutants revealed major defects in virion assembly and release in HEK 293T and HeLa cells and even larger defects in infectivity. Our data identify the clasp motif as a fundamental contributor to CA-CTD interactions necessary for efficient retroviral infection. IMPORTANCE The C-terminal domain of the capsid protein of many retroviruses has been shown to be critical for virion assembly and maturation, but the functions of this region of M-PMV are uncertain. We show that a short "clasp" motif in the capsid domain of the M-PMV Gag protein plays a key role in M-PMV virion assembly, genome packaging, and infectivity.


Asunto(s)
Proteínas de la Cápside/metabolismo , Productos del Gen gag/química , Productos del Gen gag/metabolismo , Virus del Mono Mason-Pfizer/fisiología , Virión/metabolismo , Ensamble de Virus , Secuencias de Aminoácidos , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Microscopía por Crioelectrón , Productos del Gen gag/genética , Genoma Viral , Células HEK293 , Células HeLa , Humanos , Virus del Mono Mason-Pfizer/genética , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Dominios Proteicos , ARN Viral/genética , ARN Viral/metabolismo , Empaquetamiento del Genoma Viral
10.
Science ; 370(6513)2020 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-33033190

RESUMEN

During the first half of the viral life cycle, HIV-1 reverse transcribes its RNA genome and integrates the double-stranded DNA copy into a host cell chromosome. Despite progress in characterizing and inhibiting these processes, in situ mechanistic and structural studies remain challenging. This is because these operations are executed by individual viral preintegration complexes deep within cells. We therefore reconstituted and imaged the early stages of HIV-1 replication in a cell-free system. HIV-1 cores released from permeabilized virions supported efficient, capsid-dependent endogenous reverse transcription to produce double-stranded DNA genomes, which sometimes looped out from ruptured capsid walls. Concerted integration of both viral DNA ends into a target plasmid then proceeded in a cell extract-dependent reaction. This reconstituted system uncovers the role of the capsid in templating replication.


Asunto(s)
Cápside/fisiología , VIH-1/fisiología , Integración Viral , Replicación Viral , Sistema Libre de Células , Humanos
11.
J Virol ; 95(2)2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-33115869

RESUMEN

The human immunodeficiency virus type 1 (HIV-1) capsid (CA) protein forms a conical lattice around the viral ribonucleoprotein complex (vRNP) consisting of a dimeric viral genome and associated proteins, together constituting the viral core. Upon entry into target cells, the viral core undergoes a process termed uncoating, during which CA molecules are shed from the lattice. Although the timing and degree of uncoating are important for reverse transcription and integration, the molecular basis of this phenomenon remains unclear. Using complementary approaches, we assessed the impact of core destabilization on the intrinsic stability of the CA lattice in vitro and fates of viral core components in infected cells. We found that substitutions in CA can impact the intrinsic stability of the CA lattice in vitro in the absence of vRNPs, which mirrored findings from an assessment of CA stability in virions. Altering CA stability tended to increase the propensity to form morphologically aberrant particles, in which the vRNPs were mislocalized between the CA lattice and the viral lipid envelope. Importantly, destabilization of the CA lattice led to premature dissociation of CA from vRNPs in target cells, which was accompanied by proteasomal-independent losses of the viral genome and integrase enzyme. Overall, our studies show that the CA lattice protects the vRNP from untimely degradation in target cells and provide the mechanistic basis of how CA stability influences reverse transcription.IMPORTANCE The human immunodeficiency virus type 1 (HIV-1) capsid (CA) protein forms a conical lattice around the viral RNA genome and the associated viral enzymes and proteins, together constituting the viral core. Upon infection of a new cell, viral cores are released into the cytoplasm where they undergo a process termed "uncoating," i.e., shedding of CA molecules from the conical lattice. Although proper and timely uncoating has been shown to be important for reverse transcription, the molecular mechanisms that link these two events remain poorly understood. In this study, we show that destabilization of the CA lattice leads to premature dissociation of CA from viral cores, which exposes the viral genome and the integrase enzyme for degradation in target cells. Thus, our studies demonstrate that the CA lattice protects the viral ribonucleoprotein complexes from untimely degradation in target cells and provide the first causal link between how CA stability affects reverse transcription.


Asunto(s)
Cápside/metabolismo , Genoma Viral , Integrasa de VIH/metabolismo , VIH-1/fisiología , Desencapsidación Viral , Animales , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Línea Celular , Cricetinae , Humanos , Mutación , ARN Viral/metabolismo , Transcripción Reversa , Proteínas del Núcleo Viral/metabolismo , Virión/genética , Virión/metabolismo
12.
J Biol Chem ; 295(45): 15183-15195, 2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-32788212

RESUMEN

To enter a cell and establish infection, HIV must first fuse its lipid envelope with the host cell plasma membrane. Whereas the process of HIV membrane fusion can be tracked by fluorescence microscopy, the 3D configuration of proteins and lipids at intermediate steps can only be resolved with cryo-electron tomography (cryoET). However, cryoET of whole cells is technically difficult. To overcome this problem, we have adapted giant plasma membrane vesicles (or blebs) from native cell membranes expressing appropriate receptors as targets for fusion with HIV envelope glycoprotein-expressing pseudovirus particles with and without Serinc host restriction factors. The fusion behavior of these particles was probed by TIRF microscopy on bleb-derived supported membranes. Timed snapshots of fusion of the same particles with blebs were examined by cryo-ET. The combination of these methods allowed us to characterize the structures of various intermediates on the fusion pathway and showed that when Serinc3 or Serinc5 (but not Serinc2) were present, later fusion products were more prevalent, suggesting that Serinc3/5 act at multiple steps to prevent progression to full fusion. In addition, the antifungal amphotericin B reversed Serinc restriction, presumably by intercalation into the fusing membranes. Our results provide a highly detailed view of Serinc restriction of HIV-cell membrane fusion and thus extend current structural and functional information on Serinc as a lipid-binding protein.


Asunto(s)
Membrana Celular/metabolismo , Microscopía por Crioelectrón , VIH-1/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Línea Celular , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Fusión de Membrana , Microscopía Fluorescente , Productos del Gen env del Virus de la Inmunodeficiencia Humana/metabolismo
13.
Nat Commun ; 11(1): 1307, 2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-32161265

RESUMEN

The tripartite-motif protein, TRIM5α, is an innate immune sensor that potently restricts retrovirus infection by binding to human immunodeficiency virus capsids. Higher-ordered oligomerization of this protein forms hexagonally patterned structures that wrap around the viral capsid, despite an anomalously low affinity for the capsid protein (CA). Several studies suggest TRIM5α oligomerizes into a lattice with a symmetry and spacing that matches the underlying capsid, to compensate for the weak affinity, yet little is known about how these lattices form. Using a combination of computational simulations and electron cryo-tomography imaging, we reveal the dynamical mechanisms by which these lattices self-assemble. Constrained diffusion allows the lattice to reorganize, whereas defects form on highly curved capsid surfaces to alleviate strain and lattice symmetry mismatches. Statistical analysis localizes the TRIM5α binding interface at or near the CypA binding loop of CA. These simulations elucidate the molecular-scale mechanisms of viral capsid cellular compartmentalization by TRIM5α.


Asunto(s)
Cápside/metabolismo , VIH-1/inmunología , Multimerización de Proteína/inmunología , Proteínas de Motivos Tripartitos/metabolismo , Cápside/química , Cápside/inmunología , Química Computacional , Microscopía por Crioelectrón , Cristalografía por Rayos X , Resistencia a la Enfermedad , Tomografía con Microscopio Electrónico , Proteína p24 del Núcleo del VIH/química , Proteína p24 del Núcleo del VIH/inmunología , Proteína p24 del Núcleo del VIH/metabolismo , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Humanos , Inmunidad Innata , Simulación de Dinámica Molecular , Dominios Proteicos , Proteínas de Motivos Tripartitos/química , Proteínas de Motivos Tripartitos/inmunología
14.
J Virol ; 94(5)2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31801870

RESUMEN

The assembly of an orthoretrovirus such as HIV-1 requires the coordinated functioning of multiple biochemical activities of the viral Gag protein. These activities include membrane targeting, lattice formation, packaging of the RNA genome, and recruitment of cellular cofactors that modulate assembly. In most previous studies, these Gag activities have been investigated individually, which provided somewhat limited insight into how they functionally integrate during the assembly process. Here, we report the development of a biochemical reconstitution system that allowed us to investigate how Gag lattice formation, RNA binding, and the assembly cofactor inositol hexakisphosphate (IP6) synergize to generate immature virus particles in vitro The results identify an important rate-limiting step in assembly and reveal new insights into how RNA and IP6 promote immature Gag lattice formation. The immature virus-like particles can be converted into mature capsid-like particles by the simple addition of viral protease, suggesting that it is possible in principle to fully biochemically reconstitute the sequential processes of HIV-1 assembly and maturation from purified components.IMPORTANCE Assembly and maturation are essential steps in the replication of orthoretroviruses such as HIV-1 and are proven therapeutic targets. These processes require the coordinated functioning of the viral Gag protein's multiple biochemical activities. We describe here the development of an experimental system that allows an integrative analysis of how Gag's multiple functionalities cooperate to generate a retrovirus particle. Our current studies help to illuminate how Gag synergizes the formation of the virus compartment with RNA binding and how these activities are modulated by the small molecule IP6. Further development and use of this system should lead to a more comprehensive understanding of the molecular mechanisms of HIV-1 assembly and maturation and may provide new insights for the development of antiretroviral drugs.


Asunto(s)
VIH-1/genética , VIH-1/fisiología , Ensamble de Virus/genética , Ensamble de Virus/fisiología , Cápside/metabolismo , Humanos , Microscopía Electrónica , Modelos Moleculares , Ácido Fítico , Virión/metabolismo , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/química , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo
15.
Sci Adv ; 5(11): eaaw3631, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31807695

RESUMEN

TRIM5α is a restriction factor that senses incoming retrovirus cores through an unprecedented mechanism of nonself recognition. TRIM5α assembles a hexagonal lattice that avidly binds the capsid shell, which surrounds and protects the virus core. The extent to which the TRIM lattice can cover the capsid and how TRIM5α directly contacts the capsid surface have not been established. Here, we apply cryo-electron tomography and subtomogram averaging to determine structures of TRIM5α bound to recombinant HIV-1 capsid assemblies. Our data support a mechanism of hierarchical assembly, in which a limited number of basal interaction modes are successively organized in increasingly higher-order structures that culminate in a TRIM5α cage surrounding a retroviral capsid. We further propose that cage formation explains the mechanism of restriction and provides the structural context that links capsid recognition to ubiquitin-dependent processes that disable the retrovirus.


Asunto(s)
Cápside/química , VIH-1/química , Proteínas de Motivos Tripartitos/química , Ubiquitina-Proteína Ligasas/química , Ensamble de Virus , Factores de Restricción Antivirales , Cápside/metabolismo , Cápside/ultraestructura , Microscopía por Crioelectrón , VIH-1/fisiología , VIH-1/ultraestructura , Humanos , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
16.
J Virol ; 93(24)2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31578292

RESUMEN

Interferons (IFNs) induce the expression of interferon-stimulated genes (ISGs), many of which are responsible for the cellular antiviral state in which the replication of numerous viruses is blocked. How the majority of individual ISGs inhibit the replication of particular viruses is unknown. We conducted a loss-of-function screen to identify genes required for the activity of alpha interferon (IFN-α) against vesicular stomatitis virus, Indiana serotype (VSVIND), a prototype negative-strand RNA virus. Our screen revealed that TRIM69, a member of the tripartite motif (TRIM) family of proteins, is a VSVIND inhibitor. TRIM69 potently inhibited VSVIND replication through a previously undescribed transcriptional inhibition mechanism. Specifically, TRIM69 physically associates with the VSVIND phosphoprotein (P), requiring a specific peptide target sequence encoded therein. P is a cofactor for the viral polymerase and is required for viral RNA synthesis, as well as the assembly of replication compartments. By targeting P, TRIM69 inhibits pioneer transcription of the incoming virion-associated minus-strand RNA, thereby preventing the synthesis of viral mRNAs, and consequently impedes all downstream events in the VSVIND replication cycle. Unlike some TRIM proteins, TRIM69 does not inhibit viral replication by inducing degradation of target viral proteins. Rather, higher-order TRIM69 multimerization is required for its antiviral activity, suggesting that TRIM69 functions by sequestration or anatomical disruption of the viral machinery required for VSVIND RNA synthesis.IMPORTANCE Interferons are important antiviral cytokines that work by inducing hundreds of host genes whose products inhibit the replication of many viruses. While the antiviral activity of interferon has long been known, the identities and mechanisms of action of most interferon-induced antiviral proteins remain to be discovered. We identified gene products that are important for the antiviral activity of interferon against vesicular stomatitis virus (VSV), a model virus that whose genome consists of a single RNA molecule with negative-sense polarity. We found that a particular antiviral protein, TRIM69, functions by a previously undescribed molecular mechanism. Specifically, TRIM69 interacts with and inhibits the function of a particular phosphoprotein (P) component of the viral transcription machinery, preventing the synthesis of viral messenger RNAs.


Asunto(s)
Interferón-alfa/farmacología , Proteínas de Motivos Tripartitos/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Virus de la Estomatitis Vesicular Indiana/efectos de los fármacos , Vesiculovirus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Antivirales/farmacología , Línea Celular , Citocinas/farmacología , Humanos , Modelos Moleculares , Fosfoproteínas/genética , Conformación Proteica , Dominios Proteicos , ARN Mensajero/metabolismo , ARN Viral/biosíntesis , Proteínas de Motivos Tripartitos/química , Ubiquitina-Proteína Ligasas/química , Estomatitis Vesicular/virología , Virus de la Estomatitis Vesicular Indiana/genética , Vesiculovirus/genética , Proteínas Virales
17.
Nat Rev Microbiol ; 17(9): 546-556, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31312031

RESUMEN

Mammalian cells express a variety of innate immune proteins - known as restriction factors - which defend against invading retroviruses such as HIV-1. Two members of the tripartite motif protein family - TRIM5α and TRIMCyp - were identified in 2004 as restriction factors that recognize and inactivate the capsid shell that surrounds and protects the incoming retroviral core. Research on these TRIM5 proteins has uncovered a novel mode of non-self recognition that protects against cross-species transmission of retroviruses. Our developing understanding of the mechanism of TRIM5 restriction underscores the concept that core uncoating and reverse transcription of the viral genome are coordinated processes rather than discrete steps of the post-entry pathway of retrovirus replication. In this Review, we provide an overview of the current state of knowledge of the molecular mechanism of TRIM5-mediated restriction, highlight recent advances and discuss implications for the development of capsid-targeted antiviral therapeutics.


Asunto(s)
VIH-1/inmunología , Interacciones Huésped-Patógeno , Inmunidad Innata , Factores Inmunológicos/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Factores de Restricción Antivirales , VIH-1/crecimiento & desarrollo , Humanos , Transcripción Reversa/inmunología , Desencapsidación Viral/inmunología
18.
Curr Opin Virol ; 36: 47-55, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31185449

RESUMEN

During retrovirus maturation, cleavage of the precursor structural Gag polyprotein by the viral protease induces architectural rearrangement of the virus particle from an immature into a mature, infectious form. The structural rearrangement encapsidates the viral RNA genome in a fullerene capsid, producing a diffusible viral core that can initiate infection upon entry into the cytoplasm of a host cell. Maturation is an important therapeutic window against HIV-1. In this review, we highlight recent breakthroughs in understanding of the structures of retroviral immature and mature capsid lattices that define the boundary conditions of maturation and provide novel insights on capsid transformation. We also discuss emerging insights on encapsidation of the viral genome in the mature capsid, as well as remaining questions for further study.


Asunto(s)
Cápside/química , Genoma Viral , Retroviridae/fisiología , Ensamble de Virus , Cápside/fisiología , Proteínas de la Cápside/genética , Proteínas de la Cápside/fisiología , VIH-1/genética , VIH-1/fisiología , Modelos Moleculares , Péptido Hidrolasas/genética , ARN Viral , Retroviridae/enzimología , Retroviridae/genética , Virión/genética , Virión/fisiología
19.
J Mol Biol ; 431(15): 2900-2909, 2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-31125568

RESUMEN

MuRF1 (TRIM63) is a RING-type E3 ubiquitin ligase with a predicted tripartite TRIM fold. TRIM proteins rely upon the correct placement of an N-terminal RING domain, with respect to C-terminal, specific substrate-binding domains. The TRIM domain organization is orchestrated by a central helical domain that forms an antiparallel coiled-coil motif and mediates the dimerization of the fold. MuRF1 has a reduced TRIM composition characterized by a lack of specific substrate binding domains, but contains in its helical domain a conserved sequence motif termed COS-box that has been speculated to fold independently into an α-hairpin. These characteristics had led to question whether MuRF1 adopts a canonical TRIM fold. Using a combination of electron paramagnetic resonance, on spin-labeled protein, and disulfide crosslinking, we show that TRIM63 follows the structural conservation of the TRIM dimerization domain, observed in other proteins. We also show that the COS-box motif folds back onto the dimerization coiled-coil motif, predictably forming a four-helical bundle at the center of the protein and emulating the architecture of canonical TRIMs.


Asunto(s)
Proteínas Musculares/química , Proteínas de Motivos Tripartitos/química , Ubiquitina-Proteína Ligasas/química , Cristalografía por Rayos X , Espectroscopía de Resonancia por Spin del Electrón , Humanos , Modelos Moleculares , Conformación Proteica , Conformación Proteica en Hélice alfa , Dominios Proteicos , Pliegue de Proteína , Multimerización de Proteína
20.
Cell Host Microbe ; 24(6): 751-753, 2018 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-30543772

RESUMEN

The restriction factor TRIM5α recognizes incoming retroviral capsids and blocks virus replication. In this issue of Cell Host & Microbe, Fletcher et al. (2018) show how the TRIM5α RING domain mediates formation of various ubiquitin conjugates that differentially regulate TRIM5α turnover, inhibition of viral reverse transcription, and innate immune activation.


Asunto(s)
Proteínas Portadoras , Retroviridae , Cápside , Ubiquitina , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA