Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Viruses ; 15(5)2023 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-37243184

RESUMO

African swine fever virus (ASFV) encodes more than 150 proteins, most of them of unknown function. We used a high-throughput proteomic analysis to elucidate the interactome of four ASFV proteins, which potentially mediate a critical step of the infection cycle, the fusion and endosomal exit of the virions. Using affinity purification and mass spectrometry, we were able to identify potential interacting partners for those ASFV proteins P34, E199L, MGF360-15R and E248R. Representative molecular pathways for these proteins were intracellular and Golgi vesicle transport, endoplasmic reticulum organization, lipid biosynthesis, and cholesterol metabolism. Rab geranyl geranylation emerged as a significant hit, and also Rab proteins, which are crucial regulators of the endocytic pathway and interactors of both p34 and E199L. Rab proteins co-ordinate a tight regulation of the endocytic pathway that is necessary for ASFV infection. Moreover, several interactors were proteins involved in the molecular exchange at ER membrane contacts. These ASFV fusion proteins shared interacting partners, suggesting potential common functions. Membrane trafficking and lipid metabolism were important categories, as we found significant interactions with several enzymes of the lipid metabolism. These targets were confirmed using specific inhibitors with antiviral effect in cell lines and macrophages.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Suínos , Animais , Vírus da Febre Suína Africana/fisiologia , Proteínas Virais de Fusão/metabolismo , Proteômica , Linhagem Celular
3.
Int J Mol Sci ; 23(7)2022 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-35408808

RESUMO

Microtubule targeting agents (MTAs) have been exploited mainly as anti-cancer drugs because of their impact on cellular division and angiogenesis. Additionally, microtubules (MTs) are key structures for intracellular transport, which is frequently hijacked during viral infection. We have analyzed the antiviral activity of clinically used MTAs in the infection of DNA and RNA viruses, including SARS-CoV-2, to find that MT destabilizer agents show a higher impact than stabilizers in the viral infections tested, and FDA-approved anti-helminthic benzimidazoles were among the most active compounds. In order to understand the reasons for the observed antiviral activity, we studied the impact of these compounds in motor proteins-mediated intracellular transport. To do so, we used labeled peptide tools, finding that clinically available MTAs impaired the movement linked to MT motors in living cells. However, their effect on viral infection lacked a clear correlation to their effect in motor-mediated transport, denoting the complex use of the cytoskeleton by viruses. Finally, we further delved into the molecular mechanism of action of Mebendazole by combining biochemical and structural studies to obtain crystallographic high-resolution information of the Mebendazole-tubulin complex, which provided insights into the mechanisms of differential toxicity between helminths and mammalians.


Assuntos
Tratamento Farmacológico da COVID-19 , Mebendazol , Animais , Antivirais/farmacologia , Mamíferos , Mebendazol/farmacologia , Microtúbulos , SARS-CoV-2 , Tubulina (Proteína)
4.
Antiviral Res ; 186: 105011, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33428961

RESUMO

Despite the efforts to develop new treatments against Ebola virus (EBOV) there is currently no antiviral drug licensed to treat patients with Ebola virus disease (EVD). Therefore, there is still an urgent need to find new drugs to fight against EBOV. In order to do this, a virtual screening was done on the druggable interaction between the EBOV glycoprotein (GP) and the host receptor NPC1 with a subsequent selection of compounds for further validation. This screening led to the identification of new small organic molecules with potent inhibitory action against EBOV infection using lentiviral EBOV-GP-pseudotype viruses. Moreover, some of these compounds have shown their ability to interfere with the intracellular cholesterol transport receptor NPC1 using an ELISA-based assay. These preliminary results pave the way to hit to lead optimization programs that lead to successful candidates.


Assuntos
Antivirais/farmacologia , Descoberta de Drogas/métodos , Proteína C1 de Niemann-Pick/metabolismo , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus/efeitos dos fármacos , Animais , Antivirais/isolamento & purificação , Chlorocebus aethiops , Células HEK293 , Células HeLa , Doença pelo Vírus Ebola/tratamento farmacológico , Humanos , Células Vero
5.
J Nanobiotechnology ; 16(1): 33, 2018 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-29602307

RESUMO

BACKGROUND: Dynein is a cytoskeletal molecular motor protein that transports cellular cargoes along microtubules. Biomimetic synthetic peptides designed to bind dynein have been shown to acquire dynamic properties such as cell accumulation and active intra- and inter-cellular motion through cell-to-cell contacts and projections to distant cells. On the basis of these properties dynein-binding peptides could be used to functionalize nanoparticles for drug delivery applications. RESULTS: Here, we show that gold nanoparticles modified with dynein-binding delivery sequences become mobile, powered by molecular motor proteins. Modified nanoparticles showed dynamic properties, such as travelling the cytosol, crossing intracellular barriers and shuttling the nuclear membrane. Furthermore, nanoparticles were transported from one cell to another through cell-to-cell contacts and quickly spread to distant cells through cell projections. CONCLUSIONS: The capacity of these motor-bound nanoparticles to spread to many cells and increasing cellular retention, thus avoiding losses and allowing lower dosage, could make them candidate carriers for drug delivery.


Assuntos
Sistemas de Liberação de Medicamentos , Dineínas/metabolismo , Nanopartículas Metálicas/química , Nanotecnologia/métodos , Sequência de Aminoácidos , Animais , Linhagem Celular , Ouro/química , Humanos , Nanopartículas Metálicas/ultraestrutura , Microtúbulos/metabolismo , Peso Molecular , Membrana Nuclear/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Ligação Proteica
6.
Viruses ; 9(9)2017 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-28841179

RESUMO

African swine fever virus (ASFV) is a large DNA virus that replicates predominantly in the cell cytoplasm and is the only member of the Asfarviridae family. The virus causes an acute haemorrhagic fever, African swine fever (ASF), in domestic pigs and wild boar resulting in the death of most infected animals. Apoptosis is induced at an early stage during virus entry or uncoating. However, ASFV encodes anti-apoptotic proteins which facilitate production of progeny virions. These anti-apoptotic proteins include A179L, a Bcl-2 family member; A224L, an inhibitor of apoptosis proteins (IAP) family member; EP153R a C-type lectin; and DP71L. The latter acts by inhibiting activation of the stress activated pro-apoptotic pathways pro-apoptotic pathways. The mechanisms by which these proteins act is summarised. ASF disease is characterised by massive apoptosis of uninfected lymphocytes which reduces the effectiveness of the immune response, contributing to virus pathogenesis. Mechanisms by which this apoptosis is induced are discussed.


Assuntos
Vírus da Febre Suína Africana/metabolismo , Vírus da Febre Suína Africana/patogenicidade , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Replicação Viral , Febre Suína Africana/imunologia , Vírus da Febre Suína Africana/genética , Animais , Apoptose/imunologia , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Sequência de Bases , Replicação do DNA , Distrofina , Genes Virais/genética , Genes bcl-2 , Lectinas Tipo C/genética , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Alinhamento de Sequência , Suínos , Proteínas Virais/genética , Proteínas Virais/metabolismo , Proteínas Estruturais Virais/genética , Vírion/metabolismo , Internalização do Vírus
7.
Viruses ; 9(5)2017 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-28489063

RESUMO

African swine fever (ASF) is a highly contagious viral disease of swine which causes high mortality, approaching 100%, in domestic pigs. ASF is caused by a large, double stranded DNA virus, ASF virus (ASFV), which replicates predominantly in the cytoplasm of macrophages and is the only member of the Asfarviridae family, genus Asfivirus. The natural hosts of this virus include wild suids and arthropod vectors of the Ornithodoros genus. The infection of ASFV in its reservoir hosts is usually asymptomatic and develops a persistent infection. In contrast, infection of domestic pigs leads to a lethal hemorrhagic fever for which there is no effective vaccine. Identification of ASFV genes involved in virulence and the characterization of mechanisms used by the virus to evade the immune response of the host are recognized as critical steps in the development of a vaccine. Moreover, the interplay of the viral products with host pathways, which are relevant for virus replication, provides the basic information needed for the identification of potential targets for the development of intervention strategies against this disease.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana/virologia , Suínos/virologia , Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/imunologia , Vírus da Febre Suína Africana/fisiologia , Vírus da Febre Suína Africana/ultraestrutura , Animais , Apoptose , Autofagia , Reservatórios de Doenças/virologia , Estresse do Retículo Endoplasmático , Febres Hemorrágicas Virais , Interações Hospedeiro-Patógeno , Ornithodoros/virologia , Sus scrofa/virologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Virulência , Internalização do Vírus , Replicação Viral
8.
Virus Res ; 223: 181-9, 2016 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-27497620

RESUMO

African swine fever virus (ASFV) is the etiological agent of a contagious and often lethal disease of domestic pigs that has significant economic consequences for the swine industry. The viral genome encodes for more than 150 genes, and only a select few of these genes have been studied in some detail. Here we report the characterization of open reading frame Ep152R that has a predicted complement control module/SCR domain. This domain is found in Vaccinia virus proteins that are involved in blocking the immune response during viral infection. A recombinant ASFV harboring a HA tagged version of the Ep152R protein was developed (ASFV-G-Ep152R-HA) and used to demonstrate that Ep152R is an early virus protein. Attempts to construct recombinant viruses having a deleted Ep152R gene were consistently unsuccessful indicating that Ep152R is an essential gene. Interestingly, analysis of host-protein interactions for Ep152R using a yeast two-hybrid screen, identified BAG6, a protein previously identified as being required for ASFV replication. Furthermore, fluorescent microscopy analysis confirms that Ep152R-BAG6 interaction actually occurs in cells infected with ASFV.


Assuntos
Vírus da Febre Suína Africana/fisiologia , Febre Suína Africana/metabolismo , Febre Suína Africana/virologia , Genes Essenciais , Chaperonas Moleculares/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Animais , Células Cultivadas , Sequência Conservada , Interações Hospedeiro-Patógeno , Macrófagos/metabolismo , Macrófagos/virologia , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Fases de Leitura Aberta , Ligação Proteica , Conformação Proteica , Mapeamento de Interação de Proteínas/métodos , Transporte Proteico , Deleção de Sequência , Suínos , Técnicas do Sistema de Duplo-Híbrido , Proteínas Virais/química , Replicação Viral
9.
PLoS One ; 11(4): e0154366, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27116236

RESUMO

The interferon-induced transmembrane (IFITM) protein family is a group of antiviral restriction factors that impair flexibility and inhibit membrane fusion at the plasma or the endosomal membrane, restricting viral progression at entry. While IFITMs are widely known to inhibit several single-stranded RNA viruses, there are limited reports available regarding their effect in double-stranded DNA viruses. In this work, we have analyzed a possible antiviral function of IFITMs against a double stranded DNA virus, the African swine fever virus (ASFV). Infection with cell-adapted ASFV isolate Ba71V is IFN sensitive and it induces IFITMs expression. Interestingly, high levels of IFITMs caused a collapse of the endosomal pathway to the perinuclear area. Given that ASFV entry is strongly dependent on endocytosis, we investigated whether IFITM expression could impair viral infection. Expression of IFITM1, 2 and 3 reduced virus infectivity in Vero cells, with IFITM2 and IFITM3 having an impact on viral entry/uncoating. The role of IFITM2 in the inhibition of ASFV in Vero cells could be related to impaired endocytosis-mediated viral entry and alterations in the cholesterol efflux, suggesting that IFITM2 is acting at the late endosome, preventing the decapsidation stage of ASFV.


Assuntos
Vírus da Febre Suína Africana/patogenicidade , Antígenos de Diferenciação/metabolismo , Interferons/metabolismo , Proteínas de Membrana/metabolismo , Internalização do Vírus , Animais , Membrana Celular/metabolismo , Chlorocebus aethiops , Colesterol/metabolismo , Endocitose , Endossomos/metabolismo , Células HEK293 , Humanos , Microscopia de Fluorescência , Vírus de RNA/metabolismo , RNA de Cadeia Dupla , Células Vero
10.
Virus Res ; 213: 219-223, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26732484

RESUMO

The modulation of the expression of caspases by viruses influences the cell survival of different cell types. Equine arteritis virus (EAV) induces apoptosis of BHK21 and Vero cell lines, but it is not known whether EAV induces apoptosis in RK13 cells, a common cell line routinely used in EAV diagnosis and research. In this study, we determined that caspase-3 expression was triggered after infection of RK13 cells with EAV in a time- and dose-dependent manner. We also detected caspase-8 and caspase-9 activation, indicating the stimulation of both extrinsic and intrinsic apoptosis pathways. Finally, we found caspase-12 activation, an indicator of endoplasmic reticulum stress-induced apoptosis. The variability observed in the apoptotic response in the different cell lines demonstrates that apoptosis depends on the distinctive sensitivity of each cell line used for investigation.


Assuntos
Apoptose , Estresse do Retículo Endoplasmático , Células Epiteliais/fisiologia , Células Epiteliais/virologia , Equartevirus/crescimento & desenvolvimento , Animais , Caspase 3/análise , Caspase 8/análise , Caspase 9/análise , Linhagem Celular , Coelhos
11.
J Virol ; 90(3): 1534-43, 2016 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-26608317

RESUMO

UNLABELLED: African swine fever virus (ASFV) is a major threat for porcine production that has been slowly spreading in Eastern Europe since its first appearance in the Caucasus in 2007. ASFV enters the cell by endocytosis and gains access to the cytosol to start replication from late endosomes and multivesicular bodies. Cholesterol associated with low-density lipoproteins entering the cell by endocytosis also follows a trafficking pathway similar to that of ASFV. Here we show that cholesterol plays an essential role in the establishment of infection as the virus traffics through the endocytic pathway. In contrast to the case for other DNA viruses, such as vaccinia virus or adenovirus 5, cholesterol efflux from endosomes is required for ASFV release/entry to the cytosol. Accumulation of cholesterol in endosomes impairs fusion, resulting in retention of virions inside endosomes. ASFV also remodels intracellular cholesterol by increasing its cellular uptake and redistributes free cholesterol to viral replication sites. Our analysis reveals that ASFV manipulates cholesterol dynamics to ensure an appropriate lipid flux to establish productive infection. IMPORTANCE: Since its appearance in the Caucasus in 2007, African swine fever (ASF) has been spreading westwards to neighboring European countries, threatening porcine production. Due to the lack of an effective vaccine, ASF control relies on early diagnosis and widespread culling of infected animals. We investigated early stages of ASFV infection to identify potential cellular targets for therapeutic intervention against ASF. The virus enters the cell by endocytosis, and soon thereafter, viral decapsidation occurs in the acid pH of late endosomes. We found that ASFV infection requires and reorganizes the cellular lipid cholesterol. ASFV requires cholesterol to exit the endosome to gain access to the cytoplasm to establish productive replication. Our results indicate that there is a differential requirement for cholesterol efflux for vaccinia virus or adenovirus 5 compared to ASFV.


Assuntos
Vírus da Febre Suína Africana/fisiologia , Colesterol/metabolismo , Endossomos/metabolismo , Endossomos/virologia , Internalização do Vírus , Animais , Chlorocebus aethiops , Concentração de Íons de Hidrogênio , Análise do Fluxo Metabólico , Células Vero
12.
Virus Res ; 200: 45-55, 2015 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-25662020

RESUMO

The main cellular target for African swine fever virus (ASFV) is the porcine macrophage. However, existing data about the early phases of infection were previously characterized in non-leukocyte cells such as Vero cells. Here, we report that ASFV enters the natural host cell using dynamin-dependent and clathrin-mediated endocytosis. This pathway is strongly pH-dependent during the first steps of infection in porcine macrophages. We investigated the effect of drugs inhibiting several endocytic pathways in macrophages and compared ASFV with vaccinia virus (VV), which apparently involves different entry pathways. The presence of cholesterol in cellular membranes was found to be essential for a productive ASFV infection while actin-dependent endocytosis and the participation of phosphoinositide-3-kinase (PI3K) activity were other cellular factors required in the process of viral entry. These findings improved our understanding of the ASFV interactions with macrophages that allow for successful viral replication.


Assuntos
Vírus da Febre Suína Africana/fisiologia , Colesterol/metabolismo , Clatrina/metabolismo , Endocitose , Macrófagos/virologia , Febre Suína Africana/enzimologia , Febre Suína Africana/metabolismo , Febre Suína Africana/fisiopatologia , Febre Suína Africana/virologia , Vírus da Febre Suína Africana/genética , Animais , Chlorocebus aethiops , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Suínos , Células Vero , Internalização do Vírus
13.
PLoS One ; 7(11): e48853, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23133661

RESUMO

Here we analyzed the dependence of African swine fever virus (ASFV) infection on the integrity of the endosomal pathway. Using confocal immunofluorescence with antibodies against viral capsid proteins, we found colocalization of incoming viral particles with early endosomes (EE) during the first minutes of infection. Conversely, viral capsid protein was not detected in acidic late endosomal compartments, multivesicular bodies (MVBs), late endosomes (LEs) or lysosomes (LY). Using an antibody against a viral inner core protein, we found colocalization of viral cores with late compartments from 30 to 60 minutes postinfection. The absence of capsid protein staining in LEs and LYs suggested that virus desencapsidation would take place at the acid pH of these organelles. In fact, inhibitors of intraluminal acidification of endosomes caused retention of viral capsid staining virions in Rab7 expressing endosomes and more importantly, severely impaired subsequent viral protein production. Endosomal acidification in the first hour after virus entry was essential for successful infection but not thereafter. In addition, altering the balance of phosphoinositides (PIs) which are responsible of the maintenance of the endocytic pathway impaired ASFV infection. Early infection steps were dependent on the production of phosphatidylinositol 3-phosphate (PtdIns3P) which is involved in EE maturation and multivesicular body (MVB) biogenesis and on the interconversion of PtdIns3P to phosphatidylinositol 3, 5-biphosphate (PtdIns(3,5)P(2)). Likewise, GTPase Rab7 activity should remain intact, as well as processes related to LE compartment physiology, which are crucial during early infection. Our data demonstrate that the EE and LE compartments and the integrity of the endosomal maturation pathway orchestrated by Rab proteins and PIs play a central role during early stages of ASFV infection.


Assuntos
Vírus da Febre Suína Africana/metabolismo , Febre Suína Africana/virologia , Endossomos/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Células COS , Capsídeo/metabolismo , Chlorocebus aethiops , Citoplasma/metabolismo , Endocitose , Concentração de Íons de Hidrogênio , Microscopia Confocal/métodos , Fosfatos de Fosfatidilinositol/química , Suínos , Células Vero , proteínas de unión al GTP Rab7
14.
J Virol ; 84(20): 10792-801, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20686048

RESUMO

Several viruses target the microtubular motor system in early stages of the viral life cycle. African swine fever virus (ASFV) protein p54 hijacks the microtubule-dependent transport by interaction with a dynein light chain (DYNLL1/DLC8). This was shown to be a high-affinity interaction, and the residues gradually disappearing were mapped on DLC8 to define a putative p54 binding surface by nuclear magnetic resonance (NMR) spectroscopy. The potential of short peptides targeting the binding domain to disrupt this high-affinity protein-protein interaction was assayed, and a short peptide sequence was shown to bind and compete with viral protein binding to dynein. Given the complexity and number of proteins involved in cellular transport, the prevention of this viral-DLC8 interaction might not be relevant for successful viral infection. Thus, we tested the capacity of these peptides to interfere with viral infection by disrupting dynein interaction with viral p54. Using this approach, we report on short peptides that inhibit viral growth.


Assuntos
Vírus da Febre Suína Africana/efeitos dos fármacos , Antivirais/farmacologia , Dineínas/efeitos dos fármacos , Peptídeos/farmacologia , Proteínas Estruturais Virais/efeitos dos fármacos , Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/patogenicidade , Vírus da Febre Suína Africana/fisiologia , Sequência de Aminoácidos , Animais , Antivirais/química , Ligação Competitiva , Chlorocebus aethiops , Dineínas/química , Dineínas/genética , Dineínas/fisiologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Técnicas In Vitro , Modelos Moleculares , Proteínas Motores Moleculares/química , Proteínas Motores Moleculares/efeitos dos fármacos , Proteínas Motores Moleculares/genética , Proteínas Motores Moleculares/fisiologia , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Peptídeos/química , Peptídeos/genética , Domínios e Motivos de Interação entre Proteínas , Homologia de Sequência de Aminoácidos , Sus scrofa , Células Vero , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/fisiologia
15.
FEBS Lett ; 582(23-24): 3275-80, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18775702

RESUMO

Heterogeneous nuclear ribonucleoprotein K (hnRNP-K) was identified as interacting cellular protein with the abundant immediate early protein p30 from African swine fever virus (ASFV) in a macrophage cDNA library screening. The interacting regions of hnRNP-K with p30 were established within residues 35-197, which represent KH1 and KH2 domains responsible for RNA binding. Colocalization of hnRNP-K and p30 was observed mainly in the nucleus, but not in the cytoplasm of infected cells and infection modified hnRNP-K subcellular distribution and decreased the incorporation of 5-fluorouridine into nascent RNA. Since similar effects were observed in cells transiently expressing p30, this interaction provides new insights into p30 function and could represent a possible additional mechanism by which ASFV downregulates host cell mRNA translation.


Assuntos
Febre Suína Africana/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/metabolismo , Fosfoproteínas/metabolismo , Proteínas Virais/metabolismo , Animais , Núcleo Celular/metabolismo , Chlorocebus aethiops , Biblioteca Gênica , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/genética , Macrófagos/metabolismo , Fosfoproteínas/genética , Biossíntese de Proteínas , RNA Mensageiro/genética , Suínos/metabolismo , Suínos/virologia , Técnicas do Sistema de Duplo-Híbrido , Células Vero , Proteínas Virais/genética
16.
Virology ; 375(2): 561-72, 2008 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-18329683

RESUMO

Several large DNA viruses encode Bcl-2 protein homologues involved in the regulation of the cellular apoptosis cascade. This regulation often involves the interaction of these viral proteins with diverse cellular Bcl-2 family members. We have identified the specific interactions of A179L, an African swine fever virus (ASFV) Bcl-2 homologue, with the active forms of the porcine BH3-only Bid protein (truncated Bid p13 and p15). Transient expression of ASFV A179L gene in Vero cells prevented apoptosis induced by these active forms of Bid protein. Interestingly, A179L protein was able to interact, also with the main core Bcl-2 proapoptotic proteins Bax and Bak, and with several BH3-only proteins with selective binding restrictions for full length Bid and Noxa. These results suggest a fine regulation for A179L action in the suppression of apoptosis in infected cells which is essential for efficient virus replication.


Assuntos
Vírus da Febre Suína Africana/fisiologia , Febre Suína Africana/virologia , Proteínas Reguladoras de Apoptose/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Virais/fisiologia , Sequência de Aminoácidos , Animais , Apoptose/fisiologia , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/genética , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Linhagem Celular , Regulação para Baixo , Genes bcl-2 , Dados de Sequência Molecular , Ligação Proteica/fisiologia , Suínos
17.
Virus Genes ; 35(1): 81-5, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16964554

RESUMO

Post-translational modification of proteins is a key regulatory event in many cellular processes. African swine fever virus (ASFV) is a large DNA virus that contains about 150 open reading frames (ORF) which encode for more than 150 polypeptides, most of them without assigned function. Two-dimensional gel electrophoresis (2DE) followed by Post-Source Decay Matrix-Assisted Laser Desorption/Ionization Mass Spectrometry (PSD-MALDI-MS) revealed that ASFV protein pE120R, essential for virus transport from assembly sites to plasma membranes, is acetylated at the N-terminal Ala residue during infection. To our knowledge, this is the first acetylated ASFV protein described and this modification might be relevant to ASFV life cycle since many viruses use the acetylation signaling pathway as a primary target for viral proteins after infection.


Assuntos
Acetiltransferases/metabolismo , Vírus da Febre Suína Africana/metabolismo , Proteínas de Ligação a DNA/metabolismo , Processamento de Proteína Pós-Traducional , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Proteínas Estruturais Virais/metabolismo , Acetilação , Sequência de Aminoácidos , Dados de Sequência Molecular
18.
Virology ; 350(1): 1-14, 2006 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-16490226

RESUMO

Many stages of African swine fever virus infection have not yet been studied in detail. To track the behavior of African swine fever virus (ASFV) in the infected cells in real time, we produced an infectious recombinant ASFV (B54GFP-2) that expresses and incorporates into the virus particle a chimera of the p54 envelope protein fused to the enhanced green fluorescent protein (EGFP). The incorporation of the fusion protein into the virus particle was confirmed immunologically and it was determined that p54-EGFP was fully functional by confirmation that the recombinant virus made normal-sized plaques and presented similar growth curves to the wild-type virus. The tagged virus was visualized as individual fluorescent particles during the first stages of infection and allowed to visualize the infection progression in living cells through the viral life cycle by confocal microscopy. In this work, diverse potential applications of B54GFP-2 to study different aspects of ASFV infection are shown. By using this recombinant virus it was possible to determine the trajectory and speed of intracellular virus movement. Additionally, we have been able to visualize for first time the ASFV factory formation dynamics and the cytophatic effect of the virus in live infected cells. Finally, we have analyzed virus progression along the infection cycle and infected cell death as time-lapse animations.


Assuntos
Vírus da Febre Suína Africana/isolamento & purificação , Vírus da Febre Suína Africana/fisiologia , Proteínas de Fluorescência Verde/análise , Proteínas Estruturais Virais/metabolismo , Vírus da Febre Suína Africana/química , Animais , Sobrevivência Celular , Chlorocebus aethiops , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Células Vero , Proteínas Estruturais Virais/genética
19.
FEBS Lett ; 569(1-3): 224-8, 2004 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-15225638

RESUMO

A specific interaction of ASFV p54 protein with 8 kDa light chain cytoplasmic dynein (DLC8) has been previously characterized and this interaction is critical during virus internalization and transport to factory sites. During early phases of infection, the virus induces the initiation of apoptosis triggering activation of caspase-9 and -3. To analyze the role of the structural protein p54 in apoptosis, transient expression experiments of p54 in Vero cells were carried out which resulted in effector caspase-3 activation and apoptosis. Interestingly, p54 mutants, lacking the 13 aa dynein-binding motif lose caspase activation ability and pro-death function of p54. This is the first reported ASFV protein which induces apoptosis.


Assuntos
Vírus da Febre Suína Africana , Apoptose/efeitos dos fármacos , Proteínas Virais/farmacologia , Proteínas Estruturais Virais/farmacologia , Vírus da Febre Suína Africana/genética , Animais , Sequência de Bases , Caspases/metabolismo , Chlorocebus aethiops , Primers do DNA , Ativação Enzimática , Genes Virais , Mitocôndrias/virologia , Reação em Cadeia da Polimerase , Deleção de Sequência , Transfecção , Células Vero , Proteínas Virais/genética , Proteínas Estruturais Virais/genética
20.
Proteomics ; 4(7): 2037-46, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15221765

RESUMO

Changes in gene expression are produced in cells as a consequence of virus infections. In the present work, we used proteomic technology to globally examine African swine fever virus (ASFV)-infected Vero cells searching for infection-associated proteins in order to determine target proteins for pathogenesis studies. We studied the alterations in cellular protein profile after ASFV infection by two-dimensional electrophoresis, identifying the modified cellular proteins by matrix-assisted laser desorption/ionization peptide mass fingerprinting. A total of twelve different over-expressed cellular proteins were unambiguously identified. The most significant changes were in redox-related proteins, nucleoside diphosphate kinases, heat shock proteins, members of the Ran-Gppnhp-Ranbd1 complex and apolipoproteins. These cellular protein modifications could represent distinct roles during infection related to apoptosis and transcriptional modulation mechanisms. The present study constitutes the first attempt to understand the dynamics of ASFV-host cell interactions by proteomics.


Assuntos
Vírus da Febre Suína Africana/metabolismo , Peptídeos/química , Proteômica/métodos , Sequência de Aminoácidos , Animais , Chlorocebus aethiops , Eletroforese em Gel Bidimensional , Dados de Sequência Molecular , Oxirredução , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas/química , Coloração pela Prata , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA