Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
PLoS Pathog ; 20(9): e1012473, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39235994

RESUMEN

Viroporins are small, hydrophobic viral proteins that modify cellular membranes to form tiny pores for influx of ions and small molecules. Previously, viroporins were identified exclusively in vertebrate viruses. Recent studies have shown that both plant-infecting positive-sense single-stranded (+ss) and negative-sense single-stranded (-ss) RNA viruses also encode functional viroporins. These seminal discoveries not only advance our understanding of the distribution and evolution of viroporins, but also open up a new field of plant virus research.


Asunto(s)
Enfermedades de las Plantas , Virus de Plantas , Virus de Plantas/genética , Virus de Plantas/fisiología , Enfermedades de las Plantas/virología , Proteínas Viroporinas/genética , Proteínas Viroporinas/metabolismo , Plantas/virología
2.
Front Cell Infect Microbiol ; 14: 1383917, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39119292

RESUMEN

Introduction: Oxysterol-binding protein (OSBP) is known for its crucial role in lipid transport, facilitating cholesterol exchange between the Golgi apparatus and endoplasmic reticulum membranes. Despite its established function in cellular processes, its involvement in coronavirus replication remains unclear. Methods: In this study, we investigated the role of OSBP in coronavirus replication and explored the potential of a novel OSBP-binding compound, ZJ-1, as an antiviral agent against coronaviruses, including SARS-CoV-2. We utilized a combination of biochemical and cellular assays to elucidate the interactions between OSBP and SARS-CoV-2 non-structural proteins (Nsps) and other viral proteins. Results: Our findings demonstrate that OSBP positively regulates coronavirus replication. Moreover, treatment with ZJ-1 resulted in reduced OSBP levels and exhibited potent antiviral effects against multiple coronaviruses. Through our investigation, we identified specific interactions between OSBP and SARS-CoV-2 Nsps, particularly Nsp3, Nsp4, and Nsp6, which are involved in double-membrane vesicle formation-a crucial step in viral replication. Additionally, we observed that Nsp3 a.a.1-1363, Nsp4, and Nsp6 target vesicle-associated membrane protein (VAMP)-associated protein B (VAP-B), which anchors OSBP to the ER membrane. Interestingly, the interaction between OSBP and VAP-B is disrupted by Nsp3 a.a.1-1363 and partially impaired by Nsp6. Furthermore, we identified SARS-CoV-2 orf7a, orf7b, and orf3a as additional OSBP targets, with OSBP contributing to their stabilization. Conclusion: Our study highlights the significance of OSBP in coronavirus replication and identifies it as a promising target for the development of antiviral therapies against SARS-CoV-2 and other coronaviruses. These findings underscore the potential of OSBP-targeted interventions in combating coronavirus infections.


Asunto(s)
Antivirales , Receptores de Esteroides , SARS-CoV-2 , Proteínas no Estructurales Virales , Replicación Viral , Replicación Viral/efectos de los fármacos , Humanos , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Antivirales/farmacología , Receptores de Esteroides/metabolismo , Proteínas no Estructurales Virales/metabolismo , COVID-19/virología , COVID-19/metabolismo , Chlorocebus aethiops , Células Vero , Proteínas Virales/metabolismo , Células HEK293 , Animales , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Proteínas Viroporinas/metabolismo , Proteasas Similares a la Papaína de Coronavirus/metabolismo , Unión Proteica
3.
Viruses ; 16(8)2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-39205188

RESUMEN

The interaction between SARS-CoV PDZ-binding motifs (PBMs) and cellular PDZs is responsible for virus virulence. The PBM sequence present in the 3a and envelope (E) proteins of SARS-CoV can potentially bind to over 400 cellular proteins containing PDZ domains. The role of SARS-CoV 3a and E proteins was studied. SARS-CoVs, in which 3a-PBM and E-PMB have been deleted (3a-PBM-/E-PBM-), reduced their titer around one logarithmic unit but still were viable. In addition, the absence of the E-PBM and the replacement of 3a-PBM with that of E did not allow the rescue of SARS-CoV. E protein PBM was necessary for virulence, activating p38-MAPK through the interaction with Syntenin-1 PDZ domain. However, the presence or absence of the homologous motif in the 3a protein, which does not bind to Syntenin-1, did not affect virus pathogenicity. Mutagenesis analysis and in silico modeling were performed to study the extension of the PBM of the SARS-CoV E protein. Alanine and glycine scanning was performed revealing a pair of amino acids necessary for optimum virus replication. The binding of E protein with the PDZ2 domain of the Syntenin-1 homodimer induced conformational changes in both PDZ domains 1 and 2 of the dimer.


Asunto(s)
Proteínas de la Envoltura de Coronavirus , Dominios PDZ , Unión Proteica , SARS-CoV-2 , Humanos , Virulencia , SARS-CoV-2/patogenicidad , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiología , Proteínas de la Envoltura de Coronavirus/metabolismo , Proteínas de la Envoltura de Coronavirus/genética , Animales , Proteínas Viroporinas/metabolismo , Proteínas Viroporinas/genética , COVID-19/virología , Chlorocebus aethiops , Células Vero , Secuencias de Aminoácidos , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , Replicación Viral
4.
J Med Virol ; 96(7): e29752, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38949191

RESUMEN

Antiviral signaling, immune response and cell metabolism are dysregulated by SARS-CoV-2, the causative agent of COVID-19. Here, we show that SARS-CoV-2 accessory proteins ORF3a, ORF9b, ORF9c and ORF10 induce a significant mitochondrial and metabolic reprogramming in A549 lung epithelial cells. While ORF9b, ORF9c and ORF10 induced largely overlapping transcriptomes, ORF3a induced a distinct transcriptome, including the downregulation of numerous genes with critical roles in mitochondrial function and morphology. On the other hand, all four ORFs altered mitochondrial dynamics and function, but only ORF3a and ORF9c induced a marked alteration in mitochondrial cristae structure. Genome-Scale Metabolic Models identified both metabolic flux reprogramming features both shared across all accessory proteins and specific for each accessory protein. Notably, a downregulated amino acid metabolism was observed in ORF9b, ORF9c and ORF10, while an upregulated lipid metabolism was distinctly induced by ORF3a. These findings reveal metabolic dependencies and vulnerabilities prompted by SARS-CoV-2 accessory proteins that may be exploited to identify new targets for intervention.


Asunto(s)
COVID-19 , Mitocondrias , SARS-CoV-2 , Proteínas Virales , Humanos , Células A549 , COVID-19/metabolismo , COVID-19/virología , COVID-19/patología , Mitocondrias/metabolismo , Sistemas de Lectura Abierta , SARS-CoV-2/genética , Transcriptoma , Proteínas Virales/genética , Proteínas Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Viroporinas/metabolismo
5.
J Virol ; 98(8): e0023124, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-38980063

RESUMEN

African swine fever virus (ASFV) is the causative agent of a contagious disease affecting wild and domestic swine. The function of B169L protein, as a potential integral structural membrane protein, remains to be experimentally characterized. Using state-of-the-art bioinformatics tools, we confirm here earlier predictions indicating the presence of an integral membrane helical hairpin, and further suggest anchoring of this protein to the ER membrane, with both terminal ends facing the lumen of the organelle. Our evolutionary analysis confirmed the importance of purifying selection in the preservation of the identified domains during the evolution of B169L in nature. Also, we address the possible function of this hairpin transmembrane domain (HTMD) as a class IIA viroporin. Expression of GFP fusion proteins in the absence of a signal peptide supported B169L insertion into the ER as a Type III membrane protein and the formation of oligomers therein. Overlapping peptides that spanned the B169L HTMD were reconstituted into ER-like membranes and the adopted structures analyzed by infrared spectroscopy. Consistent with the predictions, B169L transmembrane sequences adopted α-helical conformations in lipid bilayers. Moreover, single vesicle permeability assays demonstrated the assembly of lytic pores in ER-like membranes by B169L transmembrane helices, a capacity confirmed by ion-channel activity measurements in planar bilayers. Emphasizing the relevance of these observations, pore-forming activities were not observed in the case of transmembrane helices derived from EP84R, another ASFV protein predicted to anchor to membranes through a α-helical HTMD. Overall, our results support predictions of viroporin-like function for the B169L HTMD.IMPORTANCEAfrican swine fever (ASF), a devastating disease affecting domestic swine, is widely spread in Eurasia, producing significant economic problems in the pork industry. Approaches to prevent/cure the disease are mainly restricted to the limited information concerning the role of most of the genes encoded by the large (160-170 kba) virus genome. In this report, we present the experimental data on the functional characterization of the African swine fever virus (ASFV) gene B169L. Data presented here indicates that the B169L gene encodes for an essential membrane-associated protein with a viroporin function.


Asunto(s)
Virus de la Fiebre Porcina Africana , Virus de la Fiebre Porcina Africana/genética , Virus de la Fiebre Porcina Africana/metabolismo , Animales , Porcinos , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Dominios Proteicos , Proteínas Viroporinas/metabolismo , Proteínas Viroporinas/genética , Fiebre Porcina Africana/virología , Fiebre Porcina Africana/metabolismo , Proteínas Virales/metabolismo , Proteínas Virales/genética , Proteínas Virales/química , Secuencia de Aminoácidos
6.
Int J Mol Sci ; 25(12)2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38928009

RESUMEN

The COVID-19 pandemic was caused by infection with Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), which may lead to serious respiratory, vascular and neurological dysfunctions. The SARS-CoV-2 envelope protein (E protein) is a structural viroporin able to form ion channels in cell membranes, which is critical for viral replication. However, its effects in primary neurons have not been addressed. Here we used fluorescence microscopy and calcium imaging to study SARS-CoV-2 viroporin E localization and the effects on neuron damage and intracellular Ca2+ homeostasis in a model of rat hippocampal neurons aged in vitro. We found that the E protein quickly enters hippocampal neurons and colocalizes with the endoplasmic reticulum (ER) in both short-term (6-8 days in vitro, DIV) and long-term (20-22 DIV) cultures resembling young and aged neurons, respectively. Strikingly, E protein treatment induces apoptosis in aged neurons but not in young neurons. The E protein induces variable increases in cytosolic Ca2+ concentration in hippocampal neurons. Ca2+ responses to the E protein are due to Ca2+ release from intracellular stores at the ER. Moreover, E protein-induced Ca2+ release is very small in young neurons and increases dramatically in aged neurons, consistent with the enhanced Ca2+ store content in aged neurons. We conclude that the SARS-CoV-2 E protein quickly translocates to ER endomembranes of rat hippocampal neurons where it releases Ca2+, probably acting like a viroporin, thus producing Ca2+ store depletion and neuron apoptosis in aged neurons and likely contributing to neurological damage in COVID-19 patients.


Asunto(s)
Calcio , Retículo Endoplásmico , Hipocampo , Neuronas , SARS-CoV-2 , Animales , Ratas , Neuronas/metabolismo , Neuronas/virología , Neuronas/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/citología , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de la Envoltura de Coronavirus/metabolismo , COVID-19/virología , COVID-19/metabolismo , Células Cultivadas , Apoptosis/efectos de los fármacos , Cultivo Primario de Células , Muerte Celular/efectos de los fármacos , Proteínas Viroporinas/metabolismo
7.
Biochimie ; 225: 1-9, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38703943

RESUMEN

Inhibition of autophagy is one of the hallmarks of the SARS-CoV-2 infection. Recently it was reported that SARS-CoV-2 protein ORF3a inhibits fusion of autophagosomes with lysosomes via interaction with VPS39 thus preventing binding of homotypic fusion and protein sorting (HOPS) complex to RAB7 GTPase. Here we report that myelin basic protein (MBP), a major structural component of the myelin sheath, binds ORF3a and is colocalized with it in mammalian cells. Co-expression of MBP with ORF3a restores autophagy in mammalian cells, inhibited by viral protein. Our data suggest that basic charge of MBP drives suppression of ORF3a-induced autophagy inhibition as its deaminated variants lost ability to bind ORF3a and counteract autophagy blockade. These results together with our recent findings, indicating that MBP interacts with structural components of the vesicle transport machinery-synaptosomal-associated protein 23 (SNAP23), vesicle-associated membrane protein 3 (VAMP3) and Sec1/Munc18-1 family members, may suggest protective role of the MBP in terms of the maintaining of protein traffic and autophagosome-lysosome fusion machinery in oligodendrocytes during SARS-CoV-2 infection. Finally, our data may indicate that deimination of MBP observed in the patients with multiple sclerosis (MS) may contribute to the previously reported worser outcomes of COVID-19 and increase of post-COVID-19 neurologic symptoms in patients with MS.


Asunto(s)
Autofagia , Proteína Básica de Mielina , SARS-CoV-2 , Proteínas Viroporinas , Humanos , SARS-CoV-2/metabolismo , Proteína Básica de Mielina/metabolismo , Proteínas Viroporinas/metabolismo , COVID-19/metabolismo , COVID-19/virología , Animales , Autofagosomas/metabolismo , Células HEK293 , Unión Proteica , Chlorocebus aethiops
8.
Proc Natl Acad Sci U S A ; 121(21): e2401748121, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38739789

RESUMEN

Potyviridae, the largest family of plant RNA viruses, includes many important pathogens that significantly reduce the yields of many crops worldwide. In this study, we report that the 6-kilodalton peptide 1 (6K1), one of the least characterized potyviral proteins, is an endoplasmic reticulum-localized protein. AI-assisted structure modeling and biochemical assays suggest that 6K1 forms pentamers with a central hydrophobic tunnel, can increase the cell membrane permeability of Escherichia coli and Nicotiana benthamiana, and can conduct potassium in Saccharomyces cerevisiae. An infectivity assay showed that viral proliferation is inhibited by mutations that affect 6K1 multimerization. Moreover, the 6K1 or its homologous 7K proteins from other viruses of the Potyviridae family also have the ability to increase cell membrane permeability and transmembrane potassium conductance. Taken together, these data reveal that 6K1 and its homologous 7K proteins function as viroporins in viral infected cells.


Asunto(s)
Nicotiana , Nicotiana/virología , Nicotiana/metabolismo , Potyviridae/genética , Potyviridae/metabolismo , Proteínas Virales/metabolismo , Proteínas Virales/genética , Permeabilidad de la Membrana Celular , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas Viroporinas/metabolismo , Proteínas Viroporinas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Virus de Plantas/genética , Virus de Plantas/fisiología , Enfermedades de las Plantas/virología , Potasio/metabolismo
9.
Viruses ; 16(3)2024 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-38543711

RESUMEN

Viruses have a wide repertoire of molecular strategies that focus on their replication or the facilitation of different stages of the viral cycle. One of these strategies is mediated by the activity of viroporins, which are multifunctional viral proteins that, upon oligomerization, exhibit ion channel properties with mild ion selectivity. Viroporins facilitate multiple processes, such as the regulation of immune response and inflammasome activation through the induction of pore formation in various cell organelle membranes to facilitate the escape of ions and the alteration of intracellular homeostasis. Viroporins target diverse membranes (such as the cellular membrane), endoplasmic reticulum, and mitochondria. Cumulative data regarding the importance of mitochondria function in multiple processes, such as cellular metabolism, energy production, calcium homeostasis, apoptosis, and mitophagy, have been reported. The direct or indirect interaction of viroporins with mitochondria and how this interaction affects the functioning of mitochondrial cells in the innate immunity of host cells against viruses remains unclear. A better understanding of the viroporin-mitochondria interactions will provide insights into their role in affecting host immune signaling through the mitochondria. Thus, in this review, we mainly focus on descriptions of viroporins and studies that have provided insights into the role of viroporins in hijacked mitochondria.


Asunto(s)
Proteínas Viroporinas , Virus , Proteínas Viroporinas/metabolismo , Proteínas Virales/metabolismo , Canales Iónicos/metabolismo , Inmunidad Innata
10.
Curr Pharm Des ; 30(12): 912-920, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38482627

RESUMEN

The discussion has revolved around the derivatives of amino acids and peptides containing carbocycles and their potential antiviral activity in vitro against influenza A, hepatitis C viruses, and coronavirus. Studies conducted on cell cultures reveal that aminoadamantane amino acid derivatives exhibit the capacity to hinder the replication of viruses containing viroporins. Furthermore, certain compounds demonstrate potent virucidal activity with respect to influenza A/H5N1 and hepatitis C virus particles. A conceptual framework for viroporin inhibitors has been introduced, incorporating carbocyclic motifs as membranotropic carriers in the structure, alongside a functional segment comprised of amino acids and peptides. These components correspond to the interaction with the inner surface of the channel's pore or another target protein.


Asunto(s)
Aminoácidos , Antivirales , Péptidos , Antivirales/farmacología , Antivirales/química , Antivirales/síntesis química , Humanos , Péptidos/farmacología , Péptidos/química , Péptidos/síntesis química , Aminoácidos/química , Aminoácidos/farmacología , Animales , Hepacivirus/efectos de los fármacos , Proteínas Viroporinas/antagonistas & inhibidores , Proteínas Viroporinas/metabolismo , Proteínas Viroporinas/química , Virus de la Influenza A/efectos de los fármacos
11.
mBio ; 15(1): e0303023, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38078754

RESUMEN

IMPORTANCE: The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has tragically claimed millions of lives through coronavirus disease 2019 (COVID-19), and there remains a critical gap in our understanding of the precise molecular mechanisms responsible for the associated fatality. One key viral factor of interest is the SARS-CoV-2 ORF3a protein, which has been identified as a potent inducer of host cellular proinflammatory responses capable of triggering the catastrophic cytokine storm, a primary contributor to COVID-19-related deaths. Moreover, ORF3a, much like the spike protein, exhibits a propensity for frequent mutations, with certain variants linked to the severity of COVID-19. Our previous research unveiled two distinct types of ORF3a mutant proteins, categorized by their subcellular localizations, setting the stage for a comparative investigation into the functional and mechanistic disparities between these two types of ORF3a variants. Given the clinical significance and functional implications of the natural ORF3a mutations, the findings of this study promise to provide invaluable insights into the potential roles undertaken by these mutant ORF3a proteins in the pathogenesis of COVID-19.


Asunto(s)
COVID-19 , Retículo Endoplásmico , SARS-CoV-2 , Proteínas Viroporinas , Humanos , COVID-19/virología , Síndrome de Liberación de Citoquinas/patología , Síndrome de Liberación de Citoquinas/virología , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Degradación Asociada con el Retículo Endoplásmico , Proteínas Mutantes , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Proteínas Viroporinas/genética , Proteínas Viroporinas/metabolismo
12.
Viruses ; 15(11)2023 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-38005906

RESUMEN

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has caused a global pandemic of Coronavirus Disease 2019 (COVID-19). Excessive inflammation is a hallmark of severe COVID-19, and several proteins encoded in the SARS-CoV-2 genome are capable of stimulating inflammatory pathways. Among these, the accessory protein open reading frame 3a (ORF3a) has been implicated in COVID-19 pathology. Here we investigated the roles of ORF3a in binding to TNF receptor-associated factor (TRAF) proteins and inducing nuclear factor kappa B (NF-κB) activation. X-ray crystallography and a fluorescence polarization assay revealed low-affinity binding between an ORF3a N-terminal peptide and TRAFs, and a dual-luciferase assay demonstrated NF-κB activation by ORF3a. Nonetheless, mutation of the N-terminal TRAF-binding sequence PIQAS in ORF3a did not significantly diminish NF-κB activation in our assay. Our results thus suggest that the SARS-CoV-2 protein may activate NF-κB through alternative mechanisms.


Asunto(s)
COVID-19 , FN-kappa B , Proteínas Viroporinas , Humanos , COVID-19/metabolismo , COVID-19/virología , FN-kappa B/metabolismo , Unión Proteica , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Proteínas Viroporinas/metabolismo
13.
J Virol ; 97(10): e0042623, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37830820

RESUMEN

IMPORTANCE: Severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2), the virus responsible for coronavirus disease 2019 (COVID-19), has caused a global public health crisis. The E protein, a structural protein found in this virus particle, is also known to be a viroporin. As such, it forms oligomeric ion channels or pores in the host cell membrane. However, the relationship between these two functions is poorly understood. In this study, we showed that the roles of E protein in virus particle and viroporin formation are distinct. This study contributes to the development of drugs that inhibit SARS-CoV-2 virus particle formation. Additionally, we designed a highly sensitive and high-throughput virus-like particle detection system using the HiBiT tag, which is a useful tool for studying the release of SARS-CoV-2.


Asunto(s)
Proteínas de la Envoltura de Coronavirus , SARS-CoV-2 , Humanos , COVID-19 , Lisosomas/metabolismo , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/metabolismo , Proteínas Viroporinas/metabolismo , Proteínas de la Envoltura de Coronavirus/metabolismo , Secuencias de Aminoácidos , Liberación del Virus
14.
J Struct Biol ; 215(3): 108013, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37586469

RESUMEN

Viral proteins interact with lipid membranes during various stages in the viral life cycle to propagate infection. p7 is an ion channel forming protein of Hepatitis C virus (HCV) that participates in viral assembly. Studies show that it has close ties to lipid metabolism in the cell and anionic phosphatidylserine (PS) lipids are suggested to be key for its permeabilizing function, but the mechanism of its interaction with the lipid environment is largely unknown. To begin unraveling the molecular processes of the protein, we evaluated the impact of lipid environment on the binding and insertion mechanism of p7 prior to channel formation and viral assembly using molecular dynamics simulations. It is seen that p7 is sensitive to its lipid environment and results in different remodeling patterns in membranes. Helix 1 (H1) is especially important for peptide insertion, with deeper entry taking place when the membrane contains phosphatidylserine (PS). Helix 2 (H2) and the adjacent loop connecting to Helix 3 (H3) prompts recruitment of phosphatidylethanolamine (PE) lipids to the protein binding site in membrane models with lower surface charge. This work provides perspectives on the interplay between protein-lipid dynamics and membrane composition, and insights on membrane reorganization in mechanisms of disease.


Asunto(s)
Fosfatidilserinas , Proteínas Viroporinas , Proteínas Viroporinas/metabolismo , Fosfatidilserinas/metabolismo , Proteínas Virales/química , Hepacivirus/química , Hepacivirus/metabolismo , Simulación de Dinámica Molecular
15.
PLoS Pathog ; 19(7): e1011217, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37494374

RESUMEN

Many enveloped viruses enter host cells by fusing with acidic endosomes. The fusion activity of multiple viral envelope glycoproteins does not generally affect viral membrane permeability. However, fusion induced by the Lassa virus (LASV) glycoprotein complex (GPc) is always preceded by an increase in viral membrane permeability and the ensuing acidification of the virion interior. Here, systematic investigation of this LASV fusion phenotype using single pseudovirus tracking in live cells reveals that the change in membrane barrier function is associated with the fusogenic conformational reorganization of GPc. We show that a small-molecule fusion inhibitor or mutations that impair viral fusion by interfering with GPc refolding into the post-fusion structure prevent the increase in membrane permeability. We find that the increase in virion membrane permeability occurs early during endosomal maturation and is facilitated by virus-cell contact. This increase is observed using diverse arenavirus glycoproteins, whether presented on lentivirus-based pseudoviruses or arenavirus-like particles, and in multiple different cell types. Collectively, these results suggest that conformational changes in GPc triggered by low pH and cell factor binding are responsible for virion membrane permeabilization and acidification of the virion core prior to fusion. We propose that this viroporin-like activity may augment viral fusion and/or post-fusion steps of infection, including ribonucleoprotein release into the cytoplasm.


Asunto(s)
Arenavirus , Arenavirus/genética , Proteínas Viroporinas/metabolismo , Glicoproteínas/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Virus Lassa , Internalización del Virus
16.
mBio ; 14(4): e0074923, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37477430

RESUMEN

The human cytomegalovirus (HCMV) US12 gene family contributes to virus-host interactions by regulating the virus' cell tropism and its evasion of host innate immune responses. US21, one of the 10 US12 genes (US12-US21), is a descendant of a captured cellular transmembrane BAX inhibitor motif-containing gene. It encodes a 7TMD endoplasmic reticulum (ER)-resident viroporin (pUS21) capable of reducing the Ca2+ content of ER stores, which, in turn, protects cells against apoptosis. Since regulation of Ca2+ homeostasis affects a broad range of cellular responses, including cell motility, we investigated whether pUS21 might also interfere with this cytobiological consequence of Ca2+ signaling. Indeed, deletion of the US21 gene impaired the ability of HCMV-infected cells to migrate, whereas expression of US21 protein stimulated cell migration and adhesion, as well as focal adhesion (FA) dynamics, in a way that depended on its ability to manipulate ER Ca2+ content. Mechanistic studies revealed pUS21-mediated cell migration to involve calpain 2 activation since its inhibition prevented the viroporin's effects on cell motility. Pertinently, pUS21 expression stimulated a store-operated Ca2+ entry (SOCE) mechanism that may determine the activation of calpain 2 by promoting Ca2+ entry. Furthermore, pUS21 was observed to interact with talin-1, a calpain 2 substrate, and crucial protein component of FA complexes. A functional consequence of this interaction was confirmed by talin-1 knockdown, which abrogated the pUS21-mediated increase in cell migration. Together, these results indicate the US21-encoded viroporin to be a viral regulator of cell adhesion and migration in the context of HCMV infection. IMPORTANCE Human cytomegalovirus (HCMV) is an opportunistic pathogen that owes part of its success to the capture, duplication, and tuning of cellular genes to generate modern viral proteins which promote infection and persistence in the host by interfering with many cell biochemical and physiological pathways. The US21 viral protein provides an example of this evolutionary strategy: it is a cellular-derived calcium channel that manipulates intracellular calcium homeostasis to confer edges to HCMV replication. Here, we report on the characterization of a novel function of the US21 protein as a viral regulator of cell migration and adhesion through mechanisms involving its calcium channel activity. Characterization of HCMV multifunctional regulatory proteins, like US21, supports the better understanding of viral pathogenesis and may open avenues for the design of new antiviral strategies that exploit their functions.


Asunto(s)
Citomegalovirus , Proteínas Viroporinas , Humanos , Citomegalovirus/fisiología , Proteínas Viroporinas/metabolismo , Calpaína/genética , Calpaína/metabolismo , Talina/metabolismo , Proteínas Virales/metabolismo , Canales de Calcio/metabolismo , Movimiento Celular
17.
Virus Res ; 328: 199086, 2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36894068

RESUMEN

Coronavirus disease 2019 (COVID-19) is a global pandemic caused by SARS-CoV-2 infection. Patients with severe COVID-19 exhibit robust induction of proinflammatory cytokines, which are closely associated with the development of acute respiratory distress syndrome. However, the underlying mechanisms of the NF-κB activation mediated by SARS-CoV-2 infection remain poorly understood. Here, we screened SARS-CoV-2 genes and found that ORF3a induces proinflammatory cytokines by activating the NF-κB pathway. Moreover, we found that ORF3a interacts with IKKß and NEMO and enhances the interaction of IKKß-NEMO, thereby positively regulating NF-κB activity. Together, these results suggest ORF3a may play pivotal roles in the pathogenesis of SARS-CoV-2 and provide novel insights into the interaction between host immune responses and SARS-CoV-2 infection.


Asunto(s)
COVID-19 , FN-kappa B , SARS-CoV-2 , Proteínas Viroporinas , Humanos , COVID-19/metabolismo , Citocinas , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas , SARS-CoV-2/metabolismo , Proteínas Viroporinas/metabolismo
18.
J Cell Sci ; 136(6)2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36807531

RESUMEN

The coronavirus SARS-CoV-2, the agent of the deadly COVID-19 pandemic, is an enveloped virus propagating within the endocytic and secretory organelles of host mammalian cells. Enveloped viruses modify the ionic homeostasis of organelles to render their intra-luminal milieu permissive for viral entry, replication and egress. Here, we show that infection of Vero E6 cells with the delta variant of the SARS-CoV-2 alkalinizes the endoplasmic reticulum (ER)-Golgi intermediate compartment (ERGIC) as well as lysosomes, mimicking the effect of inhibitors of vacuolar proton ATPases. We further show the envelope protein of SARS-CoV-2 accumulates in the ERGIC when expressed in mammalian cells and selectively dissipates the ERGIC pH. This viroporin action is prevented by mutations of Val25 but not Asn15 within the channel pore of the envelope (E) protein. We conclude that the envelope protein acts as a proton channel in the ERGIC to mitigate the acidity of this intermediate compartment. The altered pH homeostasis of the ERGIC likely contributes to the virus fitness and pathogenicity, making the E channel an attractive drug target for the treatment of COVID-19.


Asunto(s)
COVID-19 , Proteínas del Envoltorio Viral , Animales , Humanos , Proteínas del Envoltorio Viral/metabolismo , Proteínas Viroporinas/metabolismo , COVID-19/metabolismo , Protones , Pandemias , SARS-CoV-2/metabolismo , Aparato de Golgi/metabolismo , Lisosomas/metabolismo , Mamíferos/metabolismo
19.
Curr HIV Res ; 21(2): 99-108, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36809950

RESUMEN

BACKGROUND: The genetic diversity in HIV-1 genes affects viral pathogenesis in HIV-1 positive patients. Accessory genes of HIV-1, including vpu, are reported to play a critical role in HIV pathogenesis and disease progression. Vpu has a crucial role in CD4 degradation and virus release. The sequence heterogeneity in the vpu gene may affect disease progression in patients, therefore, the current study was undertaken to identify the role of vpu in patients defined as rapid progressors. OBJECTIVE: The objective of the study was to identify the viral determinants present on vpu that may be important in disease progression in rapid progressors. METHODS: Blood samples were collected from 13 rapid progressors. DNA was isolated from PBMCs and vpu was amplified using nested PCR. Both strands of the gene were sequenced using an automated DNA Sequencer. The characterization and analysis of vpu was done using various bioinformatics tools. RESULTS: The analysis revealed that all sequences had intact ORF and sequence heterogeneity was present across all sequences and distributed all over the gene. The synonymous substitutions, however, were higher than nonsynonymous substitutions. The phylogenetic tree analysis showed an evolutionary relationship with previously published Indian subtype C sequences. Comparatively, the cytoplasmic tail(77 - 86) showed the highest degree of variability in these sequences as determined by Entropy- one tool. CONCLUSION: The study showed that due to the robust nature of the protein, the biological activity of the protein was intact and sequence heterogeneity may promote disease progression in the study population.


Asunto(s)
Infecciones por VIH , Seropositividad para VIH , VIH-1 , Humanos , Secuencia de Aminoácidos , Filogenia , Progresión de la Enfermedad , Variación Genética , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/metabolismo , Proteínas Viroporinas/genética , Proteínas Viroporinas/metabolismo
20.
J Virol ; 96(18): e0071622, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-36098511

RESUMEN

Influenza A virus (IAV) assembly at the plasma membrane is orchestrated by at least five viral components, including hemagglutinin (HA), neuraminidase (NA), matrix (M1), the ion channel M2, and viral ribonucleoprotein (vRNP) complexes, although particle formation is observed with expression of only HA and/or NA. While these five viral components are expressed efficiently in primary human monocyte-derived macrophages (MDMs) upon IAV infection, this cell type does not support efficient HA-M2 association and IAV particle assembly at the plasma membrane. Both defects are specific to MDMs and can be reversed upon disruption of F-actin. However, the relationship between the two defects is unclear. Here, we examined whether M2 contributes to particle assembly in MDMs and if so, which region of M2 determines the susceptibility to the MDM-specific and actin-dependent suppression. An analysis using correlative fluorescence and scanning electron microscopy showed that an M2-deficient virus failed to form budding structures at the cell surface even after F-actin was disrupted, indicating that M2 is essential for virus particle formation at the MDM surface. Notably, proximity ligation analysis revealed that a single amino acid substitution in a Glu-Glu-Tyr sequence (residues 74 to 76) in the M2 cytoplasmic tail allowed the HA-M2 association to occur efficiently even in MDMs with intact actin cytoskeleton. This phenotype did not correlate with known phenotypes of the M2 substitution mutants regarding M1 interaction or vRNP packaging in epithelial cells. Overall, our study identified M2 as a target of MDM-specific restriction of IAV assembly, which requires the Glu-Glu-Tyr sequence in the cytoplasmic tail. IMPORTANCE Human MDMs represent a cell type that is nonpermissive to particle formation of influenza A virus (IAV). We previously showed that close proximity association between viral HA and M2 proteins is blocked in MDMs. However, whether MDMs express a restriction factor against IAV assembly or whether they lack a dependency factor promoting assembly remained unknown. In the current study, we determined that the M2 protein is necessary for particle formation in MDMs but is also a molecular target of the MDM-specific suppression of assembly. Substitutions in the M2 cytoplasmic tail alleviated the block in both the HA-M2 association and particle production in MDMs. These findings suggest that MDMs express dependency factors necessary for assembly but also express a factor(s) that inhibits HA-M2 association and particle formation. High conservation of the M2 sequence rendering the susceptibility to the assembly block highlights the potential for M2 as a target of antiviral strategies.


Asunto(s)
Ácido Glutámico , Hemaglutininas , Virus de la Influenza A , Macrófagos , Tirosina , Proteínas de la Matriz Viral , Proteínas Viroporinas , Ensamble de Virus , Actinas/metabolismo , Secuencia de Aminoácidos , Ácido Glutámico/genética , Hemaglutininas/metabolismo , Interacciones Microbiota-Huesped/genética , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/metabolismo , Macrófagos/virología , Neuraminidasa/genética , Neuraminidasa/metabolismo , Ribonucleoproteínas/genética , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/metabolismo , Proteínas Viroporinas/química , Proteínas Viroporinas/metabolismo , Ensamble de Virus/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA