Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 96(14): e0066522, 2022 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-35762760

RESUMO

Human astrovirus VA1 has been associated with neurological disease in immunocompromised patients, and its recent propagation in cell culture has opened the possibility to study its biology. Unlike classical human astroviruses, VA1 growth was found to be independent of trypsin during virus replication in vitro. In this work, we show that despite its independence on trypsin activation for cell infection, the VA1 capsid precursor protein, of 86 kDa (VP86), is processed intracellularly, and this proteolytic processing is important for astrovirus VA1 infectivity. Antibodies raised against different regions of the capsid precursor showed that the polyprotein can be processed starting at either its amino- or carboxy-terminal end, and they allowed us to identify those proteins of about 33 (VP33) and 38 (VP38) kDa constitute the core and the spike proteins of the mature infectious virus particles, respectively. The amino-terminal end of the spike protein was found to be Thr-348. Whether the protease involved in intracellular cleavage of the capsid precursor is of viral or cellular origin remains to be determined, but the cleavage is independent of caspases. Also, trypsin is able to degrade the capsid precursor but has no effect on VP33 and VP38 proteins when assembled into virus particles. These studies provide the basis for advancement of the knowledge of astrovirus VA1 cell entry and replication. IMPORTANCE Human astrovirus VA1 has been associated with neurological disease in immunocompromised patients. Its recent propagation in cell culture has facilitated the study of its biology. In this work, we show that despite the ability of this virus to grow in the absence of trypsin, a marked feature of human classical astroviruses, the capsid precursor protein of astrovirus VA1 is cleaved intracellularly to yield the mature infectious particles, formed by two polypeptides, VP33 that constitutes the core domain of the virus particle, and VP38 that forms the spike of the virus. These studies provide a platform to advance our knowledge on astrovirus VA1 cell entry and replication.


Assuntos
Infecções por Astroviridae , Proteínas do Capsídeo , Mamastrovirus , Precursores de Proteínas , Infecções por Astroviridae/virologia , Células CACO-2 , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Humanos , Espaço Intracelular/virologia , Mamastrovirus/fisiologia , Precursores de Proteínas/metabolismo , Tripsina/metabolismo
2.
Int J Mol Sci ; 22(12)2021 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-34204285

RESUMO

Pathogenic intracellular bacteria, parasites and viruses have evolved sophisticated mechanisms to manipulate mammalian host cells to serve as niches for persistence and proliferation. The intracellular lifestyles of pathogens involve the manipulation of membrane-bound organellar compartments of host cells. In this review, we described how normal structural organization and cellular functions of endosomes, endoplasmic reticulum, Golgi apparatus, mitochondria, or lipid droplets are targeted by microbial virulence mechanisms. We focus on the specific interactions of Salmonella, Legionella pneumophila, Rickettsia rickettsii, Chlamydia spp. and Mycobacterium tuberculosis representing intracellular bacterial pathogens, and of Plasmodium spp. and Toxoplasma gondii representing intracellular parasites. The replication strategies of various viruses, i.e., Influenza A virus, Poliovirus, Brome mosaic virus, Epstein-Barr Virus, Hepatitis C virus, severe acute respiratory syndrome virus (SARS), Dengue virus, Zika virus, and others are presented with focus on the specific manipulation of the organelle compartments. We compare the specific features of intracellular lifestyle and replication cycles, and highlight the communalities in mechanisms of manipulation deployed.


Assuntos
Interações Hospedeiro-Patógeno , Organelas/metabolismo , Animais , Transporte Biológico , Biomarcadores , Metabolismo Energético , Interações Hospedeiro-Parasita , Humanos , Espaço Intracelular/metabolismo , Espaço Intracelular/microbiologia , Espaço Intracelular/parasitologia , Espaço Intracelular/virologia , Organelas/microbiologia , Organelas/parasitologia , Organelas/ultraestrutura , Fagocitose
3.
PLoS Pathog ; 17(1): e1009275, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33513206

RESUMO

Filoviruses, such as the Ebola virus (EBOV) and Marburg virus (MARV), are causative agents of sporadic outbreaks of hemorrhagic fevers in humans. To infect cells, filoviruses are internalized via macropinocytosis and traffic through the endosomal pathway where host cathepsin-dependent cleavage of the viral glycoproteins occurs. Subsequently, the cleaved viral glycoprotein interacts with the late endosome/lysosome resident host protein, Niemann-Pick C1 (NPC1). This interaction is hypothesized to trigger viral and host membrane fusion, which results in the delivery of the viral genome into the cytoplasm and subsequent initiation of replication. Some studies suggest that EBOV viral particles activate signaling cascades and host-trafficking factors to promote their localization with host factors that are essential for entry. However, the mechanism through which these activating signals are initiated remains unknown. By screening a kinase inhibitor library, we found that receptor tyrosine kinase inhibitors potently block EBOV and MARV GP-dependent viral entry. Inhibitors of epidermal growth factor receptor (EGFR), tyrosine protein kinase Met (c-Met), and the insulin receptor (InsR)/insulin like growth factor 1 receptor (IGF1R) blocked filoviral GP-mediated entry and prevented growth of replicative EBOV in Vero cells. Furthermore, inhibitors of c-Met and InsR/IGF1R also blocked viral entry in macrophages, the primary targets of EBOV infection. Interestingly, while the c-Met and InsR/IGF1R inhibitors interfered with EBOV trafficking to NPC1, virus delivery to the receptor was not impaired in the presence of the EGFR inhibitor. Instead, we observed that the NPC1 positive compartments were phenotypically altered and rendered incompetent to permit viral entry. Despite their different mechanisms of action, all three RTK inhibitors tested inhibited virus-induced Akt activation, providing a possible explanation for how EBOV may activate signaling pathways during entry. In sum, these studies strongly suggest that receptor tyrosine kinases initiate signaling cascades essential for efficient post-internalization entry steps.


Assuntos
Ebolavirus/fisiologia , Doença pelo Vírus Ebola/virologia , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais , Animais , Chlorocebus aethiops , Ebolavirus/genética , Endocitose , Endossomos/metabolismo , Endossomos/virologia , Interações Hospedeiro-Patógeno , Humanos , Espaço Intracelular/virologia , Lisossomos/metabolismo , Transporte Proteico , Proteínas Tirosina Quinases/genética , Células Vero , Vírion , Internalização do Vírus , Replicação Viral
4.
Poult Sci ; 100(1): 26-38, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33357689

RESUMO

Duck enteritis virus (DEV) multifunctional tegument protein UL13 is predicted to be a conserved herpesvirus protein kinase; however, little is known about its subcellular localization signal. In this study, through transfection of 2 predicted nuclear signals of DEV UL13 fused to enhanced green fluorescent protein, 2 bipartite nuclear localization signals (NLS) were identified. We found that ivermectin blocked the NLS-mediated nuclear import of DEV UL13, showing that the nuclear localization signal of DEV UL13 is a classical importin α- and ß-dependent process. We constructed a DEV UL13 mutant strain in which the NLS of DEV UL13 was deleted to explore whether deletion of the NLS affects viral replication. Amino acids 4 to 7 and 90 to 96 were predicted to be NLSs, further proving that nuclear import occurs via a classical importin α- and ß-dependent process. We also found that the NLS of pUL13 had no effect on DEV replication in cell culture. Our study enhances the understanding of DEV pUL13. Taken together, these results provide significant information regarding the biological function of pUL13 during DEV infection.


Assuntos
Enterite , Mardivirus , Sinais de Localização Nuclear , Proteínas Quinases , Animais , Antiparasitários/farmacologia , Células Cultivadas , Patos , Enterite/fisiopatologia , Enterite/veterinária , Enterite/virologia , Espaço Intracelular/metabolismo , Espaço Intracelular/virologia , Ivermectina/farmacologia , Mardivirus/genética , Mardivirus/metabolismo , Mutação , Sinais de Localização Nuclear/efeitos dos fármacos , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética
5.
Viral Immunol ; 34(1): 27-40, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33021467

RESUMO

Henipaviruses are single-stranded RNA viruses that have recently emerged as zoonotic pathogens, capable of causing severe acute respiratory disease and encephalitis in humans. The prototypical henipaviruses, Hendra henipavirus and Nipah henipavirus, are a major health concern as they have high mortality rates and no currently approved human vaccine or drug therapy. Understanding the mechanisms of viral replication and pathogenicity is of critical importance for therapeutic developments. A novel target for such therapies is the Henipavirus Matrix (M) protein, a multifunctional protein that drives viral assembly and inhibits the innate immune response. These multifunctional attributes promote a complicated lifecycle: while viral replication occurs in the cytoplasm, M traffics to the nucleus, where it is ubiquitinated, for correct cellular targeting and virion packaging. In this study, we review the relationship between the structure and functions of M. In specific cases, the compatibility between structural accessibility and protein functionality is not always evident, and we highlight areas that require further investigation.


Assuntos
Henipavirus , Espaço Intracelular/metabolismo , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/metabolismo , Animais , Henipavirus/química , Humanos , Espaço Intracelular/virologia
6.
J Viral Hepat ; 27(3): 261-269, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31670859

RESUMO

The decay rate of hepatitis C virus (HCV)-infected cells during therapy has been used to determine the duration of treatment needed to attain a sustained virologic response, but with direct-acting anti-virals (DAA), this rate has been difficult to estimate. Here, we show that it is possible to estimate it, by simultaneously analysing the viral load and alanine aminotransferase (ALT) kinetics during combination DAA therapy. We modelled the HCV RNA and ALT serum kinetics in 26 patients with chronic HCV genotype 1b infection, under four different sofosbuvir-based combination treatments. In all patients, ALT decayed exponentially to a set point in the normal range by 1-3 weeks after initiation of therapy. The model indicates that the ALT decay rate during the first few weeks after initiation of therapy reflects the death rate of infected cells, with an estimated median half-life of 2.5 days in this patient population. This information allows independent estimation of the rate of loss of intracellular replication complexes during therapy. Our model also predicts that the final ALT set point is not related to the release of ALT by dying HCV-infected cells. Using ALT data, one can separately obtain information about the rate of 'cure' of HCV-infected cells versus their rate of death, something not possible when analysing only HCV RNA data. This information can be used to compare the effects of different DAA combinations and to rationally evaluate their anti-viral effects.


Assuntos
Antivirais/uso terapêutico , Hepacivirus/efeitos dos fármacos , Espaço Intracelular/virologia , Modelos Teóricos , RNA Viral/genética , Replicação Viral , Alanina Transaminase/sangue , Alanina Transaminase/metabolismo , Genótipo , Hepacivirus/fisiologia , Humanos , Resposta Viral Sustentada , Carga Viral
7.
Sci Signal ; 12(599)2019 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-31530732

RESUMO

Intracellular pathogens use complex and tightly regulated processes to enter host cells. Upon initial interactions with signaling proteins at the surface of target cells, intracellular microbes activate and co-opt specific host signaling pathways that mediate cell surface-cytosol communications to facilitate pathogen internalization. Here, we discuss the roles of host receptor tyrosine kinases (RTKs) in the establishment of productive infections by major intracellular pathogens. We evaluate the gaps in the current understanding of this process and propose a comprehensive approach for assessing the role of host cell signaling in the biology of intracellular microorganisms and viruses. We also discuss RTK-targeting strategies for the treatment of various infections.


Assuntos
Infecções Bacterianas/microbiologia , Espaço Intracelular/microbiologia , Espaço Intracelular/virologia , Receptores Proteína Tirosina Quinases/metabolismo , Viroses/virologia , Animais , Infecções Bacterianas/prevenção & controle , Membrana Celular/efeitos dos fármacos , Membrana Celular/microbiologia , Membrana Celular/virologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Inibidores de Proteínas Quinases/farmacologia , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Viroses/prevenção & controle
8.
Sci Rep ; 9(1): 8682, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31213630

RESUMO

Dengue fever is one of the most important mosquito-borne viral infections in large parts of tropical and subtropical countries and is a significant public health concern and socioeconomic burden. There is an urgent need to develop antivirals that can effectively reduce dengue virus (DENV) replication and decrease viral load. Niclosamide, an antiparasitic drug approved for human use, has been recently identified as an effective antiviral agent against a number of pH-dependent viruses, including flaviviruses. Here, we reveal that neutralization of low-pH intracellular compartments by niclosamide affects multiple steps of the DENV infectious cycle. Specifically, niclosamide-induced endosomal neutralization not only prevents viral RNA replication but also affects the maturation of DENV particles, rendering them non-infectious. We found that niclosamide-induced endosomal neutralization prevented E glycoprotein conformational changes on the virion surface of flaviviruses, resulting in the release of non-infectious immature virus particles with uncleaved pr peptide from host cells. Collectively, our findings support the potential application of niclosamide as an antiviral agent against flavivirus infection and highlight a previously uncharacterized mechanism of action of the drug.


Assuntos
Vesículas Citoplasmáticas/efeitos dos fármacos , Vírus da Dengue/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Espaço Intracelular/efeitos dos fármacos , Niclosamida/farmacologia , Animais , Antivirais/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Vesículas Citoplasmáticas/química , Vesículas Citoplasmáticas/virologia , Vírus da Dengue/genética , Vírus da Dengue/crescimento & desenvolvimento , Endossomos/química , Endossomos/virologia , Humanos , Concentração de Íons de Hidrogênio , Espaço Intracelular/química , Espaço Intracelular/virologia , Estágios do Ciclo de Vida/efeitos dos fármacos , Células Vero , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Vírion/efeitos dos fármacos , Vírion/genética , Vírion/crescimento & desenvolvimento , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
9.
Biomed Pharmacother ; 116: 108976, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31103827

RESUMO

With the development of more effective direct-acting antivirals (DAAs), dual- or triple-therapy regimens represent the major strategy used to cure chronic hepatitis C virus (HCV) infection. Thus, shorter treatment duration regimens with low burden, few adverse effects and good patient adherence are urgently needed. This study theoretically demonstrates a proof-of-concept approach for shortening therapy duration by examining HCV-infected Huh7.5 cells after treatment with a high or low fixed dose of three DAAs (simeprevir + daclatasvir + sofosbuvir) for 6-15 days. The results demonstrated that HCV-infected Huh7.5 cells achieved an ultrarapid virologic response with undetectable HCV RNA and protein and were cured after treatment with the triple-therapy regimen for 15 days. When the treatment duration was shortened, virologic relapse might occur after treatment with a low fixed dose of the three DAAs for 9 days and did occur after treatment with a low fixed dose for 6 days, although HCV was below detectable levels at the end of treatment. However, virologic relapse could be avoided with treatment of a high fixed dose of the three DAAs for 9 or 6 days. Although a virologic breakthrough occurred after an intermittent treatment regimen at the low fixed dose, the high fixed dose cured HCV-positive Huh7.5 cells with intermittent treatment. In conclusion, HCV is persistently present below detectable levels in HCV-infected Huh7.5 cells for a long time after treatment, and a shortened therapy duration is associated with an increased risk of virologic relapse, but virologic relapse or breakthrough might be avoided by treatment with a combination of more highly effective DAAs.


Assuntos
Antivirais/uso terapêutico , Hepacivirus/fisiologia , Hepatite C Crônica/tratamento farmacológico , Hepatite C Crônica/virologia , Antivirais/farmacologia , Carbamatos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sinergismo Farmacológico , Quimioterapia Combinada , Hepacivirus/efeitos dos fármacos , Humanos , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Espaço Intracelular/virologia , Pirrolidinas , Recidiva , Simeprevir/farmacologia , Simeprevir/uso terapêutico , Sofosbuvir/farmacologia , Sofosbuvir/uso terapêutico , Valina/análogos & derivados , Replicação Viral/efeitos dos fármacos
10.
J Theor Biol ; 473: 20-27, 2019 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-31004612

RESUMO

Even in the presence of a successful combination therapy stalling the progress of AIDS, developing a cure for this disease is still an open question. One of the major steps towards a cure would be to be able to eradicate latent HIV reservoirs present in patients. During the last decade, multiple findings point to the dominant role of the viral protein Tat in the establishment of latency. Here we present a mathematical study to understand the potential role of Tat inhibitors as virus-suppressing agents. For this aim, we implemented a computational model that reproduces intracellular dynamics. Simulating an HIV-infected cell and its intracellular feedback we observed that removing Tat protein from the system via inhibitors resulted in a temporary and reversible viral suppression. In contrast, we observed that compounds that interact with Tat protein and disrupt the integrated viral genome produced a more permanent viral suppression.


Assuntos
Antivirais/farmacologia , HIV/fisiologia , Modelos Biológicos , Latência Viral/efeitos dos fármacos , Produtos do Gene tat do Vírus da Imunodeficiência Humana/antagonistas & inibidores , Simulação por Computador , Regulação Viral da Expressão Gênica/efeitos dos fármacos , HIV/efeitos dos fármacos , HIV/genética , Espaço Intracelular/virologia , Fatores de Tempo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo
11.
Clin Chim Acta ; 495: 100-108, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30959045

RESUMO

Inflammasomes play a very important role in the host defense against multiple pathogenic microbes, including bacteria and viruses. Inflammasomes are multiprotein complex platforms that mediate the processing of the two most important inflammatory cytokines, pro-IL-1ß and pro-IL-18, to their active forms. The inflammasome is formed by the apoptosis-associated speck-like protein containing a CARD (ASC), procaspase-1 and a sensor protein, either a NOD-like receptor (NLR) or an absent in melanoma 2 (AIM2)-like receptor. The sensor molecule determines inflammasome specificity by detecting specific and conserved microbial products or cell stress signals. Compared with the other inflammasomes, there is much more unknown about the activation or regulation mechanisms of the AIM2 inflammasome. In this review, we will discuss these mechanisms and the specific roles of the AIM2 inflammasome in response to diverse pathogens.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Infecções/microbiologia , Infecções/virologia , Inflamassomos/metabolismo , Animais , Caspases/metabolismo , Humanos , Infecções/metabolismo , Infecções/patologia , Espaço Intracelular/metabolismo , Espaço Intracelular/microbiologia , Espaço Intracelular/virologia
12.
Adv Virus Res ; 100: 309-354, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29551141

RESUMO

Innate immunity is traditionally thought of as the first line of defense against pathogens that enter the body. It is typically characterized as a rather weak defense mechanism, designed to restrict pathogen replication until the adaptive immune response generates a tailored response and eliminates the infectious agent. However, intensive research in recent years has resulted in better understanding of innate immunity as well as the discovery of many effector proteins, revealing its numerous powerful mechanisms to defend the host. Furthermore, this research has demonstrated that it is simplistic to strictly separate adaptive and innate immune functions since these two systems often work synergistically rather than sequentially. Here, we provide a broad overview of innate pattern recognition receptors in antiviral defense, with a focus on the TRIM family, and discuss their signaling pathways and mechanisms of action with special emphasis on the intracellular antibody receptor TRIM21.


Assuntos
Imunidade Inata/imunologia , Espaço Intracelular/imunologia , Espaço Intracelular/virologia , Animais , Humanos , Imunomodulação , Moléculas com Motivos Associados a Patógenos/imunologia , Moléculas com Motivos Associados a Patógenos/metabolismo , Receptores de Reconhecimento de Padrão/imunologia , Receptores de Reconhecimento de Padrão/metabolismo , Transdução de Sinais/imunologia , Proteínas com Motivo Tripartido/química , Proteínas com Motivo Tripartido/imunologia , Proteínas com Motivo Tripartido/metabolismo , Viroses/imunologia , Viroses/virologia
13.
Viruses ; 10(4)2018 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-29561780

RESUMO

At the forefront of vector control efforts are strategies that leverage host-microbe associations to reduce vectorial capacity. The most promising of these efforts employs Wolbachia, a maternally transmitted endosymbiotic bacterium naturally found in 40% of insects. Wolbachia can spread through a population of insects while simultaneously inhibiting the replication of viruses within its host. Despite successes in using Wolbachia-transfected mosquitoes to limit dengue, Zika, and chikungunya transmission, the mechanisms behind pathogen-blocking have not been fully characterized. Firstly, we discuss how Wolbachia and viruses both require specific host-derived structures, compounds, and processes to initiate and maintain infection. There is significant overlap in these requirements, and infection with either microbe often manifests as cellular stress, which may be a key component of Wolbachia's anti-viral effect. Secondly, we discuss the current understanding of pathogen-blocking through this lens of cellular stress and develop a comprehensive view of how the lives of Wolbachia and viruses are fundamentally in conflict with each other. A thorough understanding of the genetic and cellular determinants of pathogen-blocking will significantly enhance the ability of vector control programs to deploy and maintain effective Wolbachia-mediated control measures.


Assuntos
Coinfecção , Interações Hospedeiro-Patógeno , Infecções por Rickettsiaceae/microbiologia , Simbiose , Viroses/virologia , Fenômenos Fisiológicos Virais , Wolbachia/fisiologia , Animais , Antibiose , Transporte Biológico , Resistência à Doença/genética , Resistência à Doença/imunologia , Genótipo , Humanos , Insetos/microbiologia , Insetos/virologia , Espaço Intracelular/microbiologia , Espaço Intracelular/virologia , Biossíntese de Proteínas , Interferência de RNA , Estresse Fisiológico , Virulência , Montagem de Vírus , Internalização do Vírus , Replicação Viral
15.
ACS Infect Dis ; 4(2): 88-92, 2018 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-29298032

RESUMO

There is a large, global unmet need for the development of countermeasures to combat intracellular pathogens. The development of novel antimicrobials is expensive and slow and typically focuses on selective inhibition of proteins encoded by a single pathogen, thereby providing a narrow spectrum of coverage. The repurposing of approved drugs targeting host functions required for microbial infections represents a promising alternative. This review summarizes progress and challenges in the repurposing of approved drugs as host-targeted broad-spectrum agents for the treatment of intracellular pathogens. These strategies include targeting both cellular factors required for infection by various viruses, intracellular bacteria, and/or protozoa as well as factors that modulate the host immune response to these microbial infections. The repurposed approach offers complementary means to develop therapeutics against existing and emerging intracellular microbial threats.


Assuntos
Anti-Infecciosos/farmacologia , Reposicionamento de Medicamentos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/microbiologia , Animais , Antineoplásicos/farmacologia , Humanos , Espaço Intracelular/virologia , Terapia de Alvo Molecular
16.
J Theor Biol ; 435: 229-237, 2017 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-28943403

RESUMO

Bacteriophage proteins are viruses that can significantly impact on the functioning of bacteria and can be used in phage based therapy. The functioning of Bacteriophage in the host bacteria depends on its location in those host cells. It is very important to know the subcellular location of the phage proteins in a host cell in order to understand their working mechanism. In this paper, we propose iPHLoc-ES, a prediction method for subcellular localization of bacteriophage proteins. We aim to solve two problems: discriminating between host located and non-host located phage proteins and discriminating between the locations of host located protein in a host cell (membrane or cytoplasm). To do this, we extract sets of evolutionary and structural features of phage protein and employ Support Vector Machine (SVM) as our classifier. We also use recursive feature elimination (RFE) to reduce the number of features for effective prediction. On standard dataset using standard evaluation criteria, our method significantly outperforms the state-of-the-art predictor. iPHLoc-ES is readily available to use as a standalone tool from: https://github.com/swakkhar/iPHLoc-ES/ and as a web application from: http://brl.uiu.ac.bd/iPHLoc-ES/.


Assuntos
Bacteriófagos/química , Compartimento Celular , Máquina de Vetores de Suporte/normas , Proteínas Virais/metabolismo , Evolução Molecular , Interações Hospedeiro-Patógeno , Espaço Intracelular/virologia , Modelos Biológicos , Proteínas Virais/genética
17.
J Cell Sci ; 130(1): 260-268, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27026525

RESUMO

Positive-strand RNA viruses, which can be devastating pathogens in humans, animals and plants, replicate their genomes on intracellular membranes. Here, we describe the three-dimensional ultrastructural organization of a tombusvirus replicase in yeast, a valuable model for exploring virus-host interactions. We visualized the intracellular distribution of a viral replicase protein using metal-tagging transmission electron microscopy, a highly sensitive nanotechnology whose full potential remains to be developed. These three-dimensional images show how viral replicase molecules are organized when they are incorporated into the active domains of the intracellular replication compartment. Our approach provides a means to study protein activation mechanisms in cells and to identify targets for new antiviral compounds.


Assuntos
Imageamento Tridimensional , Espaço Intracelular/virologia , RNA Viral/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Tombusvirus/fisiologia , Montagem de Vírus , Anticorpos/metabolismo , Metalotioneína/metabolismo , Modelos Biológicos , RNA de Cadeia Dupla/metabolismo , Saccharomyces cerevisiae/ultraestrutura , Saccharomyces cerevisiae/virologia , Tombusvirus/ultraestrutura , Tomografia , Replicação Viral
18.
Cell Mol Immunol ; 14(1): 14-21, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27545071

RESUMO

Originally described by the late evolutionary biologist Leigh Van Valen, the Red Queen hypothesis posits that the evolutionary arms race between hosts and their pathogens selects for discrete, genetically encoded events that lead to competitive advantages over the other species. Examples of immune evasion strategies are seen throughout the co-evolution of the mammalian immune system and pathogens, such as the enzymatic inactivation of nuclear factor-κB signaling or host translation by pathogen-encoded virulence factors. Such immunoevasive maneuvers would be expected to select for the evolution of innate immune counterstrategies. Recent advances in our understanding of host immunity and microbial pathogenesis have provided insight into a particular innate immune adaptation, termed bystander activation. Bystander activation occurs as a consequence of infected cells alerting and instructing neighboring uninfected cells to produce inflammatory mediators, either through direct cell contact or paracrine signals. Thus, bystander activation can allow the immune system to overcome the ability of pathogens to disarm immune signaling in directly infected cells. This review presents an overview of the general hallmarks of bystander activation and their emerging role in innate immunity to intracellular pathogens, as well as examples of recent mechanistic discoveries relating to the bystander activation during infection with specific pathogens relevant to human health and disease.


Assuntos
Efeito Espectador , Evasão da Resposta Imune , Imunidade Inata , Espaço Intracelular/microbiologia , Espaço Intracelular/virologia , Modelos Biológicos , Animais , Humanos , Espaço Intracelular/parasitologia
19.
PLoS One ; 11(12): e0168939, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28033419

RESUMO

The budded phenotype (BV) of the baculovirus AcMNPV has been demonstrated to have strong immunostimulatory properties that are relevant for the development of vaccines and antiviral therapies. Although the occluded phenotype (ODV) shares the main structural proteins and its genome with BV, it has been poorly studied in mammals. In this study, we assessed the capacity of ODV to induce immune responses in mice. In contrast to BVs, ODVs failed to promote the secretion of IFN-gamma, IL-6 and Il-12 and to induce antiviral activity against VSV in the short term. Furthermore, ODVs were unable to induce cellular immunity against a coadministered antigen 7 days after inoculation. By analyzing the interaction of ODVs with BMDCs, we observed that although ODVs entered the cells reaching late and acidic endosomes, they did not induce their maturation. Finally, we also analyzed if BVs and ODVs followed different routes in the cell during the infection. BVs, but not ODVs, colocalized with the protein ovalbumin in compartments with the presence of proteases. The results suggest that structural differences could be responsible for their different destinies in the dendritic cell and this could lead to a different impact on the immune response.


Assuntos
Espaço Intracelular/virologia , Nucleopoliedrovírus/fisiologia , Fenótipo , Animais , Células da Medula Óssea/citologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Endossomos/virologia , Feminino , Concentração de Íons de Hidrogênio , Camundongos , Camundongos Endogâmicos C57BL , Células Sf9 , Spodoptera
20.
Virology ; 497: 185-197, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27479465

RESUMO

Coronavirus spike proteins mediate host-cell-attachment and virus entry. Virus replication takes place within the host cell cytosol, whereas assembly and budding occur at the endoplasmic reticulum-Golgi intermediate compartment. In this study we demonstrated that the last 39 amino acid stretches of Alphacoronavirus spike cytoplasmic domains of the human coronavirus 229E, NL63, and the porcine transmissible gastroenteritis virus TGEV interact with tubulin alpha and beta chains. In addition, a partial co-localization of TGEV spike proteins with authentic host cell ß-tubulin was observed. Furthermore, drug-induced microtubule depolymerization led to changes in spike protein distribution, a reduction in the release of infectious virus particles and less amount of spike protein incorporated into virions. These data demonstrate that interaction of Alphacoronavirus spike proteins with tubulin supports S protein transport and incorporation into virus particles.


Assuntos
Infecções por Coronaviridae/metabolismo , Infecções por Coronaviridae/virologia , Coronaviridae/fisiologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Tubulina (Proteína)/metabolismo , Montagem de Vírus , Replicação Viral , Animais , Linhagem Celular , Coronaviridae/efeitos dos fármacos , Gastroenterite Suína Transmissível/metabolismo , Gastroenterite Suína Transmissível/virologia , Humanos , Espaço Intracelular/metabolismo , Espaço Intracelular/virologia , Nocodazol/farmacologia , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico , Glicoproteína da Espícula de Coronavírus/química , Suínos , Montagem de Vírus/efeitos dos fármacos , Liberação de Vírus , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...