Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 512
Filtrar
1.
J Virol ; 97(3): e0165022, 2023 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-36790205

RESUMO

Truncations of the cytoplasmic tail (CT) of entry proteins of enveloped viruses dramatically increase the infectivity of pseudoviruses (PVs) bearing these proteins. Several mechanisms have been proposed to explain this enhanced entry, including an increase in cell surface expression. However, alternative explanations have also been forwarded, and the underlying mechanisms for the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) S protein remain undetermined. Here, we show that the partial or complete deletion of the CT (residues 19 to 35) does not modify SARS-CoV-2 S protein expression on the cell surface when the S2 subunit is measured, whereas it is significantly increased when the S1 subunit is measured. We also show that the higher level of S1 in these CT-truncated S proteins reflects the decreased dissociation of the S1 subunit from the S2 subunit. In addition, we demonstrate that CT truncation further promotes S protein incorporation into PV particles, as indicated by biochemical analyses and cryo-electron microscopy. Thus, our data show that two distinct mechanisms contribute to the markedly increased infectivity of PVs carrying CT-truncated SARS-CoV-2 S proteins and help clarify the interpretation of the results of studies employing such PVs. IMPORTANCE Various forms of PVs have been used as tools to evaluate vaccine efficacy and study virus entry steps. When PV infectivity is inherently low, such as that of SARS-CoV-2, a CT-truncated version of the viral entry glycoprotein is widely used to enhance PV infectivity, but the mechanism underlying this enhanced PV infectivity has been unclear. Here, our study identified two mechanisms by which the CT truncation of the SARS-CoV-2 S protein dramatically increases PV infectivity: a reduction of S1 shedding and an increase in S protein incorporation into PV particles. An understanding of these mechanisms can clarify the mechanistic bases for the differences observed among various assays employing such PVs.


Assuntos
SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Vírion , Humanos , COVID-19/virologia , Microscopia Crioeletrônica , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidade , Glicoproteína da Espícula de Coronavírus/metabolismo , Vírion/genética , Vírion/patogenicidade , Regulação Viral da Expressão Gênica/genética
2.
Viruses ; 14(5)2022 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-35632693

RESUMO

The emergence of new SARS-CoV-2 variants and the replacement of preceding isolates have been observed through B.1.1.7, B.1.351, B.1.617.2, and B.1.1.529 lineages (corresponding to alpha, beta, delta, and omicron variants of concern (VoC), respectively). However, there is still a lack of biological evidence to which extent those VoC differ from the ancestral lineages. By exploiting human airway epithelial cell (HAEC) cultures, which closely resemble the human airway architecture and physiology, we report distinctive SARS-CoV-2 tropism in different respiratory tissues. In general, SARS-CoV-2 VoC predominantly infect and replicate in HAEC better than the progenitor USA-WA1 isolate or the BavPat1 isolate, which contains the D614G mutation, even though there is little to no difference between variants regarding their infectivity (i.e., virion-per-vRNA copy ratio). We also observe differential tissue-specific innate immunity activation between the upper and lower respiratory tissues in the presence of the virus. Our study provides better comprehension of the behavior of the different VoC in this physiologically relevant ex vivo model.


Assuntos
COVID-19 , Citocinas , Células Epiteliais , SARS-CoV-2 , COVID-19/imunologia , COVID-19/virologia , Citocinas/metabolismo , Células Epiteliais/virologia , Humanos , SARS-CoV-2/patogenicidade , Vírion/patogenicidade
3.
Life Sci ; 295: 120411, 2022 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-35181310

RESUMO

AIMS: Virus-infected host cells switch their metabolism to a more glycolytic phenotype, required for new virion synthesis and packaging. Therefore, we investigated the effect and mechanistic action of glycolytic inhibitor 2-Deoxy-d-glucose (2-DG) on virus multiplication in host cells following SARS-CoV-2 infection. MAIN METHODS: SARS-CoV-2 induced change in glycolysis was examined in Vero E6 cells. Effect of 2-DG on virus multiplication was evaluated by RT-PCR (N and RdRp genes) analysis, protein expression analysis of Nucleocapsid (N) and Spike (S) proteins and visual indication of cytopathy effect (CPE), The mass spectrometry analysis was performed to examine the 2-DG induced change in glycosylation status of receptor binding domain (RBD) in SARS-CoV-2 spike protein. KEY FINDINGS: We observed SARS-COV-2 infection induced increased glucose influx and glycolysis, resulting in selectively high accumulation of the fluorescent glucose analog, 2-NBDG in Vero E6 cells. 2-DG inhibited glycolysis, reduced virus multiplication and alleviated cells from virus-induced cytopathic effect (CPE) in SARS-CoV-2 infected cells. The progeny virions produced from 2-DG treated cells were found unglycosylated at crucial N-glycosites (N331 and N343) of the receptor-binding domain (RBD) in the spike protein, resulting in production of defective progeny virions with compromised infective potential. SIGNIFICANCE: The mechanistic study revealed that the inhibition of SARS-COV-2 multiplication is attributed to 2-DG induced glycolysis inhibition and possibly un-glycosylation of the spike protein, also. Therefore, based on its previous human trials in different types of Cancer and Herpes patients, it could be a potential molecule to study in COVID-19 patients.


Assuntos
Tratamento Farmacológico da COVID-19 , Desoxiglucose/farmacologia , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/patogenicidade , Trifosfato de Adenosina/metabolismo , Animais , Antivirais/farmacologia , COVID-19/metabolismo , COVID-19/virologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Glucose/metabolismo , Glicólise/efeitos dos fármacos , Glicosilação , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Manose/farmacologia , SARS-CoV-2/fisiologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Células Vero , Vírion/efeitos dos fármacos , Vírion/patogenicidade , Replicação Viral/efeitos dos fármacos
4.
J Cell Mol Med ; 26(1): 25-34, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34904376

RESUMO

Transmission electron microscopy has historically been indispensable for virology research, as it offers unique insight into virus function. In the past decade, as cryo-electron microscopy (cryo-EM) has matured and become more accessible, we have been able to peer into the structure of viruses at the atomic level and understand how they interact with the host cell, with drugs or with antibodies. Perhaps, there was no time in recent history where cryo-EM was more needed, as SARS-CoV-2 has spread around the globe, causing millions of deaths and almost unquantifiable economic devastation. In this concise review, we aim to mark the most important contributions of cryo-EM to understanding the structure and function of SARS-CoV-2 proteins, from surface spikes to the virus core and from virus-receptor interactions to antibody binding.


Assuntos
Enzima de Conversão de Angiotensina 2/química , Anticorpos Antivirais/química , Vacinas contra COVID-19/química , COVID-19/prevenção & controle , Receptores Virais/química , Glicoproteína da Espícula de Coronavírus/química , Enzima de Conversão de Angiotensina 2/imunologia , Enzima de Conversão de Angiotensina 2/metabolismo , Anticorpos Antivirais/biossíntese , COVID-19/imunologia , COVID-19/virologia , Vacinas contra COVID-19/administração & dosagem , Vacinas contra COVID-19/biossíntese , Microscopia Crioeletrônica , Epitopos/química , Epitopos/imunologia , Epitopos/metabolismo , Humanos , Modelos Moleculares , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Receptores Virais/imunologia , Receptores Virais/metabolismo , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/patogenicidade , SARS-CoV-2/ultraestrutura , Serina Endopeptidases/química , Serina Endopeptidases/imunologia , Serina Endopeptidases/metabolismo , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Vírion/efeitos dos fármacos , Vírion/patogenicidade , Vírion/ultraestrutura
5.
Biochim Biophys Acta Biomembr ; 1864(1): 183804, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34656553

RESUMO

Protein-lipid interactions modulate a plethora of physiopathologic processes and have been the subject of countless studies. However, these kinds of interactions in the context of viral envelopes have remained relatively unexplored, partially because the intrinsically small dimensions of the molecular systems escape to the current resolution of experimental techniques. However, coarse-grained and multiscale simulations may fill that niche, providing nearly atomistic resolution at an affordable computational price. Here we use multiscale simulations to characterize the lipid-protein interactions in the envelope of the Zika Virus, a prominent member of the Flavivirus genus. Comparisons between the viral envelope and simpler molecular systems indicate that the viral membrane is under extreme pressures and asymmetric forces. Furthermore, the dense net of protein-protein contacts established by the envelope proteins creates poorly solvated regions that destabilize the external leaflet leading to a decoupled dynamics between both membrane layers. These findings lead to the idea that the Flaviviral membrane may store a significant amount of elastic energy, playing an active role in the membrane fusion process.


Assuntos
Fusão de Membrana/genética , Lipídeos de Membrana/genética , Fagocitose/genética , Zika virus/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Humanos , Lipídeos de Membrana/metabolismo , Vírion/genética , Vírion/patogenicidade , Zika virus/patogenicidade , Infecção por Zika virus/genética , Infecção por Zika virus/virologia
6.
PLoS Pathog ; 17(12): e1010177, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34962975

RESUMO

The extracellular virion (EV) form of Orthopoxviruses is required for cell-to-cell spread and pathogenesis, and is the target of neutralizing antibodies in the protective immune response. EV have a double envelope that contains several unique proteins that are involved in its intracellular envelopment and/or subsequent infectivity. One of these, F13, is involved in both EV formation and infectivity. Here, we report that replacement of vaccinia virus F13L with the molluscum contagiosum virus homolog, MC021L, results in the production of EV particles with significantly increased levels of EV glycoproteins, which correlate with a small plaque phenotype. Using a novel fluorescence-activated virion sorting assay to isolate EV populations based on glycoprotein content we determine that EV containing either higher or lower levels of glycoproteins are less infectious, suggesting that there is an optimal concentration of glycoproteins in the outer envelope that is required for maximal infectivity of EV. This optimal glycoprotein concentration was required for lethality and induction of pathology in a cutaneous model of animal infection, but was not required for induction of a protective immune response. Therefore, our results demonstrate that there is a sensitive balance between glycoprotein incorporation, infectivity, and pathogenesis, and that manipulation of EV glycoprotein levels can produce vaccine vectors in which pathologic side effects are attenuated without a marked diminution in induction of protective immunity.


Assuntos
Glicoproteínas/metabolismo , Vírus Vaccinia/patogenicidade , Vaccinia/metabolismo , Proteínas Virais/metabolismo , Vírion/patogenicidade , Animais , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Vírus Vaccinia/metabolismo , Proteínas do Envelope Viral/metabolismo , Vírion/metabolismo
7.
Cell Host Microbe ; 29(12): 1788-1801.e6, 2021 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-34822776

RESUMO

Previous work found that the co-occurring mutations R203K/G204R on the SARS-CoV-2 nucleocapsid (N) protein are increasing in frequency among emerging variants of concern or interest. Through a combination of in silico analyses, this study demonstrates that R203K/G204R are adaptive, while large-scale phylogenetic analyses indicate that R203K/G204R associate with the emergence of the high-transmissibility SARS-CoV-2 lineage B.1.1.7. Competition experiments suggest that the 203K/204R variants possess a replication advantage over the preceding R203/G204 variants, possibly related to ribonucleocapsid (RNP) assembly. Moreover, the 203K/204R virus shows increased infectivity in human lung cells and hamsters. Accordingly, we observe a positive association between increased COVID-19 severity and sample frequency of 203K/204R. Our work suggests that the 203K/204R mutations contribute to the increased transmission and virulence of select SARS-CoV-2 variants. In addition to mutations in the spike protein, mutations in the nucleocapsid protein are important for viral spreading during the pandemic.


Assuntos
Substituição de Aminoácidos , COVID-19/patologia , Proteínas do Nucleocapsídeo de Coronavírus/genética , Genoma Viral , Mutação , SARS-CoV-2/genética , Animais , COVID-19/epidemiologia , COVID-19/virologia , Linhagem Celular , Proteínas do Nucleocapsídeo de Coronavírus/química , Proteínas do Nucleocapsídeo de Coronavírus/metabolismo , Cricetulus , Células Epiteliais/patologia , Células Epiteliais/virologia , Expressão Gênica , Aptidão Genética , Humanos , Modelos Moleculares , Mutagênese , Fosfoproteínas/química , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Filogenia , Conformação Proteica , SARS-CoV-2/classificação , SARS-CoV-2/crescimento & desenvolvimento , SARS-CoV-2/patogenicidade , Seleção Genética , Índice de Gravidade de Doença , Vírion/genética , Vírion/crescimento & desenvolvimento , Vírion/patogenicidade , Virulência , Replicação Viral
8.
STAR Protoc ; 2(4): 100940, 2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34806049

RESUMO

We describe how to use a publicly available computational model for coxsackievirus B3 (CVB3) infection that we recast as a graphical user interface (GUI). The GUI-based implementation enables non-computationalists to incorporate systems-biology modeling into their research and teaching. The model simulates the full life cycle of CVB3, including the host antiviral response, and includes 44 alterable parameters. The model simplifies some viral life cycle processes to improve interpretability and utility when performing in silico experiments. For complete details on the use and execution of this protocol, please refer to Lopacinski et al. (2021).


Assuntos
Simulação por Computador , Infecções por Coxsackievirus/virologia , Enterovirus Humano B , Biologia de Sistemas/métodos , Interface Usuário-Computador , Enterovirus Humano B/patogenicidade , Enterovirus Humano B/fisiologia , Humanos , Cinética , Software , Vírion/patogenicidade , Vírion/fisiologia
9.
Curr Opin Virol ; 51: 65-73, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34619513

RESUMO

Many icosahedral viruses including tailed bacteriophages and herpes viruses have a unique portal vertex where a dodecameric protein ring is associated with a fivefold capsid shell. While the peripheral regions of the portal ring are involved in capsid assembly, its central channel is used to transport DNA into and out of capsid during genome packaging and infection. Though the atomic structure of this highly conserved, turbine-shaped, portal is known for nearly two decades, its molecular mechanism remains a mystery. Recent high-resolution in situ structures reveal various conformational states of the portal and the asymmetric interactions between the 12-fold portal and the fivefold capsid. These lead to a valve-like mechanism for this symmetry-mismatched portal vertex that regulates DNA flow through the channel, a critical function for high fidelity assembly of an infectious virion.


Assuntos
Capsídeo/química , Vírion/química , Vírion/metabolismo , Capsídeo/metabolismo , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Vírion/patogenicidade
10.
Int J Mol Sci ; 22(20)2021 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-34681765

RESUMO

Bacillus virus Bam35 is the model Betatectivirus and member of the family Tectiviridae, which is composed of tailless, icosahedral, and membrane-containing bacteriophages. Interest in these viruses has greatly increased in recent years as they are thought to be an evolutionary link between diverse groups of prokaryotic and eukaryotic viruses. Additionally, betatectiviruses infect bacteria of the Bacillus cereus group, which are known for their applications in industry and notorious since it contains many pathogens. Here, we present the first protein-protein interactions (PPIs) network for a tectivirus-host system by studying the Bam35-Bacillus thuringiensis model using a novel approach that integrates the traditional yeast two-hybrid system and high-throughput sequencing (Y2H-HTS). We generated and thoroughly analyzed a genomic library of Bam35's host B. thuringiensis HER1410 and screened interactions with all the viral proteins using different combinations of bait-prey couples. Initial analysis of the raw data enabled the identification of over 4000 candidate interactions, which were sequentially filtered to produce 182 high-confidence interactions that were defined as part of the core virus-host interactome. Overall, host metabolism proteins and peptidases were particularly enriched within the detected interactions, distinguishing this host-phage system from the other reported host-phage PPIs. Our approach also suggested biological roles for several Bam35 proteins of unknown function, including the membrane structural protein P25, which may be a viral hub with a role in host membrane modification during viral particle morphogenesis. This work resulted in a better understanding of the Bam35-B. thuringiensis interaction at the molecular level and holds great potential for the generalization of the Y2H-HTS approach for other virus-host models.


Assuntos
Bacillus thuringiensis/virologia , Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno/fisiologia , Tectiviridae/fisiologia , Proteínas Virais/metabolismo , Bacillus thuringiensis/genética , Proteínas de Bactérias/genética , Biblioteca Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Fases de Leitura Aberta , Mapas de Interação de Proteínas , Saccharomyces cerevisiae/genética , Tectiviridae/patogenicidade , Técnicas do Sistema de Duplo-Híbrido , Proteínas Virais/genética , Vírion/patogenicidade , Vírion/fisiologia
11.
J Gen Virol ; 102(9)2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34546870

RESUMO

Tick-borne encephalitis virus (TBEV), a member of the genus Flavivirus, is common in Europe and Asia and causes a severe disease of the central nervous system. A promising approach in the development of therapy for TBEV infection is the search for small molecule antivirals targeting the flavivirus envelope protein E, particularly its ß-n-octyl-d-glucoside binding pocket (ß-OG pocket). However, experimental studies of candidate antivirals may be complicated by varying amounts and different forms of the protein E in the virus samples. Viral particles with different conformations and arrangements of the protein E are produced during the replication cycle of flaviviruses, including mature, partially mature, and immature forms, as well as subviral particles lacking genomic RNA. The immature forms are known to be abundant in the viral population. We obtained immature virion preparations of TBEV, characterized them by RT-qPCR, and assessed in vivo and in vitro infectivity of the residual mature virions in the immature virus samples. Analysis of the ß-OG pocket structure on the immature virions confirmed the possibility of binding of adamantylmethyl esters of 5-aminoisoxazole-3-carboxylic acid in the pocket. We demonstrated that the antiviral activity of these compounds in plaque reduction assay is significantly reduced in the presence of immature TBEV particles.


Assuntos
Adamantano/farmacologia , Antivirais/farmacologia , Vírus da Encefalite Transmitidos por Carrapatos/efeitos dos fármacos , Vírus da Encefalite Transmitidos por Carrapatos/fisiologia , Encefalite Transmitida por Carrapatos/virologia , Isoxazóis/farmacologia , Vírion/fisiologia , Adamantano/metabolismo , Animais , Antivirais/metabolismo , Linhagem Celular , Vírus da Encefalite Transmitidos por Carrapatos/crescimento & desenvolvimento , Vírus da Encefalite Transmitidos por Carrapatos/patogenicidade , Glucosídeos/metabolismo , Isoxazóis/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Simulação de Acoplamento Molecular , Ligação Proteica , Conformação Proteica , Suínos , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Ensaio de Placa Viral , Vírion/imunologia , Vírion/patogenicidade , Vírion/ultraestrutura
13.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34341107

RESUMO

The majority of viruses infecting hyperthermophilic archaea display unique virion architectures and are evolutionarily unrelated to viruses of bacteria and eukaryotes. The lack of relationships to other known viruses suggests that the mechanisms of virus-host interaction in Archaea are also likely to be distinct. To gain insights into archaeal virus-host interactions, we studied the life cycle of the enveloped, ∼2-µm-long Sulfolobus islandicus filamentous virus (SIFV), a member of the family Lipothrixviridae infecting a hyperthermophilic and acidophilic archaeon Saccharolobus islandicus LAL14/1. Using dual-axis electron tomography and convolutional neural network analysis, we characterize the life cycle of SIFV and show that the virions, which are nearly two times longer than the host cell diameter, are assembled in the cell cytoplasm, forming twisted virion bundles organized on a nonperfect hexagonal lattice. Remarkably, our results indicate that envelopment of the helical nucleocapsids takes place inside the cell rather than by budding as in the case of most other known enveloped viruses. The mature virions are released from the cell through large (up to 220 nm in diameter), six-sided pyramidal portals, which are built from multiple copies of a single 89-amino-acid-long viral protein gp43. The overexpression of this protein in Escherichia coli leads to pyramid formation in the bacterial membrane. Collectively, our results provide insights into the assembly and release of enveloped filamentous viruses and illuminate the evolution of virus-host interactions in Archaea.


Assuntos
Interações Hospedeiro-Patógeno/fisiologia , Lipothrixviridae/fisiologia , Lipothrixviridae/patogenicidade , Sulfolobus/virologia , Citoplasma/virologia , Tomografia com Microscopia Eletrônica , Escherichia coli/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Vírion/metabolismo , Vírion/patogenicidade
14.
Commun Biol ; 4(1): 652, 2021 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-34079056

RESUMO

Hantaan viruses (HTNVs) are zoonotic pathogens transmitted mainly by rodents and capable of infecting humans. Increasing knowledge of the human response to HTNV infection can guide the development of new preventative vaccines and therapeutic strategies. Here, we show that HTNV can infect CD8+ T cells in vivo in patients diagnosed with hemorrhagic fever with renal syndrome (HFRS). Electron microscopy-mediated tracking of the life cycle and ultrastructure of HTNV-infected CD8+ T cells in vitro showed an association between notable increases in cytoplasmic multivesicular bodies and virus production. Notably, based on a clinical cohort of 280 patients, we found that circulating HTNV-infected CD8+ T cell numbers in blood were proportional to disease severity. These results demonstrate that viral infected CD8+ T cells may be used as an adjunct marker for monitoring HFRS disease progression and that modulating T cell functions may be explored for new treatment strategies.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Vírus Hantaan/imunologia , Vírus Hantaan/patogenicidade , Febre Hemorrágica com Síndrome Renal/imunologia , Febre Hemorrágica com Síndrome Renal/virologia , Doença Aguda , Adulto , Linfócitos T CD8-Positivos/ultraestrutura , Micropartículas Derivadas de Células/ultraestrutura , Micropartículas Derivadas de Células/virologia , Citocinas/sangue , Progressão da Doença , Feminino , Vírus Hantaan/fisiologia , Febre Hemorrágica com Síndrome Renal/sangue , Humanos , Técnicas In Vitro , Masculino , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Modelos Biológicos , Vírion/imunologia , Vírion/patogenicidade , Replicação Viral
15.
Adv Virus Res ; 109: 105-134, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33934825

RESUMO

The cellular surfaceome and its residing extracellularly exposed proteins are involved in a multitude of molecular signaling processes across the viral infection cycle. Successful viral propagation, including viral entry, immune evasion, virion release and viral spread rely on dynamic molecular interactions with the surfaceome. Decoding of these viral-host surfaceome interactions using advanced technologies enabled the discovery of fundamental new functional insights into cellular and viral biology. In this review, we highlight recently developed experimental strategies, with a focus on spatial proteotyping technologies, aiding in the rational design of theranostic strategies to combat viral infections.


Assuntos
Interações entre Hospedeiro e Microrganismos , Mapeamento de Interação de Proteínas/métodos , Proteínas Virais/metabolismo , Viroses/imunologia , Interações Hospedeiro-Patógeno , Humanos , Evasão da Resposta Imune , Vírion/metabolismo , Vírion/patogenicidade , Internalização do Vírus
16.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-34001619

RESUMO

SERINC5 is a potent lentiviral restriction factor that gets incorporated into nascent virions and inhibits viral fusion and infectivity. The envelope glycoprotein (Env) is a key determinant for SERINC restriction, but many aspects of this relationship remain incompletely understood, and the mechanism of SERINC5 restriction remains unresolved. Here, we have used mutants of HIV-1 and HIV-2 to show that truncation of the Env cytoplasmic tail (ΔCT) confers complete resistance of both viruses to SERINC5 and SERINC3 restriction. Critically, fusion of HIV-1 ΔCT virus was not inhibited by SERINC5 incorporation into virions, providing a mechanism to explain how EnvCT truncation allows escape from restriction. Neutralization and inhibitor assays showed ΔCT viruses have an altered Env conformation and fusion kinetics, suggesting that EnvCT truncation dysregulates the processivity of entry, in turn allowing Env to escape targeting by SERINC5. Furthermore, HIV-1 and HIV-2 ΔCT viruses were also resistant to IFITMs, another entry-targeting family of restriction factors. Notably, while the EnvCT is essential for Env incorporation into HIV-1 virions and spreading infection in T cells, HIV-2 does not require the EnvCT. Here, we reveal a mechanism by which human lentiviruses can evade two potent Env-targeting restriction factors but show key differences in the capacity of HIV-1 and HIV-2 to exploit this. Taken together, this study provides insights into the interplay between HIV and entry-targeting restriction factors, revealing viral plasticity toward mechanisms of escape and a key role for the long lentiviral EnvCT in regulating these processes.


Assuntos
HIV-1/genética , HIV-2/genética , Glicoproteínas de Membrana/genética , Proteínas de Membrana/genética , Vírion/genética , Produtos do Gene env do Vírus da Imunodeficiência Humana/genética , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/imunologia , Linhagem Celular , Regulação da Expressão Gênica , Células HEK293 , Infecções por HIV/genética , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/imunologia , HIV-1/patogenicidade , HIV-2/imunologia , HIV-2/patogenicidade , Células HeLa , Humanos , Evasão da Resposta Imune , Glicoproteínas de Membrana/imunologia , Proteínas de Membrana/imunologia , Domínios Proteicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Transdução de Sinais , Células-Tronco , Vírion/imunologia , Vírion/patogenicidade , Internalização do Vírus , Produtos do Gene env do Vírus da Imunodeficiência Humana/imunologia
17.
Bioessays ; 43(6): e2000312, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33857328

RESUMO

Biocidal agents such as formaldehyde and glutaraldehyde are able to inactivate several coronaviruses including SARS-CoV-2. In this article, an insight into one mechanism for the inactivation of these viruses by those two agents is presented, based on analysis of previous observations during electron microscopic examination of several members of the orthocoronavirinae subfamily, including the new virus SARS-CoV-2. This inactivation is proposed to occur through Schiff base reaction-induced conformational changes in the spike glycoprotein leading to its disruption or breakage, which can prevent binding of the virus to cellular receptors. Also, a new prophylactic and therapeutic measure against SARS-CoV-2 using acetoacetate is proposed, suggesting that it could similarly break the viral spike through Schiff base reaction with lysines of the spike protein. This measure needs to be confirmed experimentally before consideration. In addition, a new line of research is proposed to help find a broad-spectrum antivirus against several members of this subfamily.


Assuntos
Desinfetantes/farmacologia , Corpos Cetônicos/farmacologia , SARS-CoV-2/efeitos dos fármacos , Glicoproteína da Espícula de Coronavírus/metabolismo , Animais , Antivirais/química , Antivirais/farmacologia , Desinfetantes/química , Formaldeído/química , Formaldeído/farmacologia , Glutaral/química , Glutaral/farmacologia , Humanos , Corpos Cetônicos/química , Corpos Cetônicos/metabolismo , Cetose/etiologia , Cetose/virologia , SARS-CoV-2/patogenicidade , Vírion/efeitos dos fármacos , Vírion/patogenicidade
18.
Virus Genes ; 57(3): 280-283, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33929643

RESUMO

Anguillid herpesvirus 1 (AngHV) is one of the vital pathogenic agents found in the wild and cultured eel populations, which has brought significant losses to eel culture industry in China. In this study, AngHV ORF95 was characterized. Bioinformatics analysis showed that ORF95 putatively encodes a structural protein that is homologous to hemagglutinin-esterase (HE) protein of infectious salmon anemia virus (ISAV). Temporal transcription and expression analysis indicated that ORF95 is a viral late gene. Subcellular localization analysis revealed that ORF95 was predominantly localized in the cytoplasm. Further, western blot analysis indicated that ORF95 is a structural protein of virion envelope. These results provide a novel basis to make further efforts to clarify the function of ORF95 in the process of AngHV infection and the possibility to use ORF95 as antigen to develop AngHV subunit vaccine.


Assuntos
Enguias/virologia , Hemaglutininas Virais/genética , Herpesviridae/genética , Fases de Leitura Aberta/genética , Proteínas Virais de Fusão/genética , Animais , Enguias/genética , Doenças dos Peixes/genética , Doenças dos Peixes/virologia , Herpesviridae/isolamento & purificação , Herpesviridae/patogenicidade , Isavirus/genética , Vírion/genética , Vírion/patogenicidade
19.
Cardiovasc Res ; 117(6): 1557-1566, 2021 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-33705542

RESUMO

AIMS: Patients with severe respiratory syndrome caused by SARS-CoV-2 undergo cardiac complications due to hyper-inflammatory conditions. Although the presence of the virus has been detected in the myocardium of infected patients, and infection of induced pluripotent cell-derived cardiomyocytes has been demonstrated, the reported expression of Angiotensin-Converting Enzyme-2 (ACE2) in cardiac stromal cells suggests that SARS-CoV-2 may determine cardiac injury by sustaining productive infection and increasing inflammation. METHODS AND RESULTS: We analysed expression of ACE2 receptor in primary human cardiac stromal cells derived from cardiospheres, using proteomics and transcriptomics before exposing them to SARS-CoV-2 in vitro. Using conventional and high sensitivity PCR methods, we measured virus release in the cellular supernatants and monitored the intracellular viral bioprocessing. We performed high-resolution imaging to show the sites of intracellular viral production and demonstrated the presence of viral particles in the cells with electron microscopy. We finally used RT-qPCR assays to detect genes linked to innate immunity and fibrotic pathways coherently regulated in cells after exposure to the virus. CONCLUSIONS: Our findings indicate that cardiac stromal cells are susceptible to SARS-CoV-2 infection and produce variable viral yields depending on the extent of cellular ACE2 receptor expression. Interestingly, these cells also evolved towards hyper-inflammatory/pro-fibrotic phenotypes independently of ACE2 levels. Thus, SARS-CoV-2 infection of myocardial stromal cells could be involved in cardiac injury and explain the high number of complications observed in severe cases of COVID-19.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/virologia , Cardiopatias/virologia , Miocárdio/enzimologia , Receptores Virais/metabolismo , SARS-CoV-2/patogenicidade , Células Estromais/virologia , Vírion/patogenicidade , Idoso , Idoso de 80 Anos ou mais , Enzima de Conversão de Angiotensina 2/genética , Animais , COVID-19/complicações , Chlorocebus aethiops , Feminino , Fibrose , Cardiopatias/enzimologia , Cardiopatias/patologia , Interações Hospedeiro-Patógeno , Humanos , Mediadores da Inflamação/metabolismo , Masculino , Pessoa de Meia-Idade , Miocárdio/ultraestrutura , Fenótipo , Receptores Virais/genética , SARS-CoV-2/ultraestrutura , Esferoides Celulares , Células Estromais/enzimologia , Células Estromais/ultraestrutura , Células Vero , Vírion/ultraestrutura
20.
Adv Colloid Interface Sci ; 290: 102400, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33713994

RESUMO

We review concepts involved in describing the chemodynamic features of nanoparticles and apply the framework to gain physicochemical insights into interactions between SARS-CoV-2 virions and airborne particulate matter (PM). Our analysis is highly pertinent given that the World Health Organisation acknowledges that SARS-CoV-2 may be transmitted by respiratory droplets, and the US Center for Disease Control and Prevention recognises that airborne transmission of SARS-CoV-2 can occur. In our theoretical treatment, the virion is assimilated to a core-shell nanoparticle, and contributions of various interaction energies to the virion-PM association (electrostatic, hydrophobic, London-van der Waals, etc.) are generically included. We review the limited available literature on the physicochemical features of the SARS-CoV-2 virion and identify knowledge gaps. Despite the lack of quantitative data, our conceptual framework qualitatively predicts that virion-PM entities are largely able to maintain equilibrium on the timescale of their diffusion towards the host cell surface. Comparison of the relevant mass transport coefficients reveals that virion biointernalization demand by alveolar host cells may be greater than the diffusive supply. Under such conditions both the free and PM-sorbed virions may contribute to the transmitted dose. This result points to the potential for PM to serve as a shuttle for delivery of virions to host cell targets. Thus, our critical review reveals that the chemodynamics of virion-PM interactions may play a crucial role in the transmission of COVID-19, and provides a sound basis for explaining reported correlations between episodes of air pollution and outbreaks of COVID-19.


Assuntos
COVID-19/transmissão , Células Epiteliais/virologia , Material Particulado/química , SARS-CoV-2/química , Vírion/química , Aerossóis , Fenômenos Biomecânicos , COVID-19/virologia , Difusão , Humanos , Interações Hidrofóbicas e Hidrofílicas , Modelos Químicos , Nanopartículas/química , Alvéolos Pulmonares/virologia , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidade , Eletricidade Estática , Vírion/metabolismo , Vírion/patogenicidade , Internalização do Vírus , Água/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...