Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochim Biophys Acta Gen Subj ; 1865(3): 129839, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33412226

RESUMO

Mitochondria are multi-functioning organelles that participate in a wide range of biologic processes from energy metabolism to cellular suicide. Mitochondria are also involved in the cellular innate immune response against microorganisms or environmental irritants, particularly in mammals. Mitochondrial-mediated innate immunity is achieved by the activation of two discrete signaling pathways, the NLR family pyrin domain-containing 3 inflammasomes and the retinoic acid-inducible gene I-like receptor pathway. In both pathways, a mitochondrial outer membrane adaptor protein, called mitochondrial antiviral signaling MAVS, and mitochondria-derived components play a key role in signal transduction. In this review, we discuss current insights regarding the fundamental phenomena of mitochondrial-related innate immune responses, and review the specific roles of various mitochondrial subcompartments in fine-tuning innate immune signaling events. We propose that specific targeting of mitochondrial functions is a potential therapeutic approach for the management of infectious diseases and autoinflammatory disorders with an excessive immune response.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata , Mitocôndrias/imunologia , Infecções por Vírus de RNA/imunologia , Vírus de RNA/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/imunologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Humanos , Inflamassomos , MicroRNAs/genética , MicroRNAs/imunologia , Mitocôndrias/genética , Mitocôndrias/virologia , Membranas Mitocondriais/imunologia , Membranas Mitocondriais/virologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Infecções por Vírus de RNA/genética , Infecções por Vírus de RNA/patologia , Infecções por Vírus de RNA/virologia , Vírus de RNA/genética , Vírus de RNA/patogenicidade , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Transdução de Sinais
2.
J Biol Chem ; 295(2): 444-457, 2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31767682

RESUMO

MicroRNAs (miRNAs) are small noncoding RNAs that suppress the expression of multiple genes and are involved in numerous biologic functions and disorders, including human diseases. Here, we report that two miRNAs, miR-302b and miR-372, target mitochondrial-mediated antiviral innate immunity by regulating mitochondrial dynamics and metabolic demand. Using human cell lines transfected with the synthetic analog of viral dsRNA, poly(I-C), or challenged with Sendai virus, we found that both miRNAs are up-regulated in the cells late after viral infection and ultimately terminate the production of type I interferons and inflammatory cytokines. We found that miR-302b and miR-372 are involved in dynamin-related protein 1 (DRP1)-dependent mitochondrial fragmentation and disrupt mitochondrial metabolism by attenuating solute carrier family 25 member 12 (SLC25A12), a member of the SLC25 family. Neutralizing the effects of the two miRNAs through specific inhibitors re-established the mitochondrial dynamics and the antiviral responses. We found that SLC25A12 contributes to regulating the antiviral response by inducing mitochondrial-related metabolite changes in the organelle. Structure-function analysis indicated that SLC25A12, as part of a prohibitin complex, associates with the mitochondrial antiviral-signaling protein in mitochondria, providing structural insight into the regulation of the mitochondrial-mediated antiviral response. Our results contribute to the understanding of how miRNAs modulate the innate immune response by altering mitochondrial dynamics and metabolic demand. Manipulating the activities of miR-302b and miR-372 may be a potential therapeutic approach to target RNA viruses.


Assuntos
MicroRNAs/metabolismo , Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Infecções por Respirovirus/metabolismo , Vírus Sendai/fisiologia , Linhagem Celular , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , MicroRNAs/imunologia , Mitocôndrias/imunologia , Mitocôndrias/virologia , Proteínas de Transporte da Membrana Mitocondrial/imunologia , Membranas Mitocondriais/imunologia , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/virologia , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/virologia , Vírus Sendai/imunologia
3.
FEBS J ; 286(8): 1543-1560, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30715798

RESUMO

Double-stranded RNA (dsRNA) is a potent proinflammatory signature of viral infection and is sensed primarily by RIG-I-like receptors (RLRs). Oligomerization of RLRs following binding to cytosolic dsRNA activates and nucleates self-assembly of the mitochondrial antiviral-signaling protein (MAVS). In the current signaling model, the caspase recruitment domains of MAVS form helical fibrils that self-propagate like prions to promote signaling complex assembly. However, there is no conclusive evidence that MAVS forms fibrils in cells or with the transmembrane anchor present. We show here with super-resolution light microscopy that MAVS activation by dsRNA induces mitochondrial membrane remodeling. Quantitative image analysis at imaging resolutions as high as 32 nm shows that in the cellular context, MAVS signaling complexes and the fibrils within them are smaller than 80 nm. The transmembrane domain of MAVS is required for its membrane remodeling, interferon signaling, and proapoptotic activities. We conclude that membrane tethering of MAVS restrains its polymerization and contributes to mitochondrial remodeling and apoptosis upon dsRNA sensing.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Interferon beta/metabolismo , Membranas Mitocondriais/metabolismo , Células 3T3/virologia , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Morte Celular/fisiologia , Citosol/fisiologia , Fibroblastos/metabolismo , Helicase IFIH1 Induzida por Interferon/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Knockout , Microscopia/métodos , Membranas Mitocondriais/virologia , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial , Domínios Proteicos , RNA de Cadeia Dupla/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Análise de Célula Única/métodos , Febre do Nilo Ocidental/metabolismo
4.
PLoS Pathog ; 15(1): e1007510, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30653614

RESUMO

Numerous plant viruses that cause significant agricultural problems are persistently transmitted by insect vectors. We wanted to see if apoptosis was involved in viral infection process in the vector. We found that a plant reovirus (rice gall dwarf virus, RGDV) induced typical apoptotic response during viral replication in the leafhopper vector and cultured vector cells, as demonstrated by mitochondrial degeneration and membrane potential decrease. Fibrillar structures formed by nonstructural protein Pns11 of RGDV targeted the outer membrane of mitochondria, likely by interaction with an apoptosis-related mitochondrial protein in virus-infected leafhopper cells or nonvector insect cells. Such association of virus-induced fibrillar structures with mitochondria clearly led to mitochondrial degeneration and membrane potential decrease, suggesting that RGDV Pns11 was the inducer of apoptotic response in insect vectors. A caspase inhibitor treatment and knockdown of caspase gene expression using RNA interference each reduced apoptosis and viral accumulation, while the knockdown of gene expression for the inhibitor of apoptosis protein improved apoptosis and viral accumulation. Thus, RGDV exploited caspase-dependent apoptotic response to promote viral infection in insect vectors. For the first time, we directly confirmed that a nonstructural protein encoded by a persistent plant virus can induce the typical apoptotic response to benefit viral transmission by insect vectors.


Assuntos
Apoptose/fisiologia , Hemípteros/virologia , Reoviridae/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Colágenos Fibrilares/metabolismo , Insetos Vetores/virologia , Insetos/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/virologia , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/virologia , Vírus de Plantas/metabolismo , Reoviridae/genética , Reoviridae/patogenicidade , Reoviridae/fisiologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral
5.
PLoS Pathog ; 14(1): e1006778, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29324904

RESUMO

A close relative of poliovirus, enterovirus 71 (EV71) is regarded as an important neurotropic virus of serious public health concern. EV71 causes Hand, Foot and Mouth Disease and has been associated with neurological complications in young children. Our limited understanding of the mechanisms involved in its neuropathogenesis has hampered the development of effective therapeutic options. Here, using a two-dimensional proteomics approach combined with mass spectrometry, we have identified a unique panel of host proteins that were differentially and dynamically modulated during EV71 infection of motor-neuron NSC-34 cells, which are found at the neuromuscular junctions where EV71 is believed to enter the central nervous system. Meta-analysis with previously published proteomics studies in neuroblastoma or muscle cell lines revealed minimal overlapping which suggests unique host-pathogen interactions in NSC-34 cells. Among the candidate proteins, we focused our attention on prohibitin (PHB), a protein that is involved in multiple cellular functions and the target of anti-cancer drug Rocaglamide (Roc-A). We demonstrated that cell surface-expressed PHB is involved in EV71 entry into neuronal cells specifically, while membrane-bound mitochondrial PHB associates with the virus replication complex and facilitates viral replication. Furthermore, Roc-A treatment of EV71-infected neuronal cells reduced significantly virus yields. However, the inhibitory effect of Roc-A on PHB in NSC-34 cells was not through blocking the CRAF/MEK/ERK pathway as previously reported. Instead, Roc-A treated NSC-34 cells had lower mitochondria-associated PHB and lower ATP levels that correlated with impaired mitochondria integrity. In vivo, EV71-infected mice treated with Roc-A survived longer than the vehicle-treated animals and had significantly lower virus loads in their spinal cord and brain, whereas virus titers in their limb muscles were comparable to controls. Together, this study uncovers PHB as the first host factor that is specifically involved in EV71 neuropathogenesis and a potential drug target to limit neurological complications.


Assuntos
Membrana Celular/virologia , Enterovirus Humano A/fisiologia , Interações Hospedeiro-Patógeno , Membranas Mitocondriais/virologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/virologia , Proteínas Repressoras/metabolismo , Animais , Antivirais/uso terapêutico , Benzofuranos/uso terapêutico , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Enterovirus Humano A/efeitos dos fármacos , Enterovirus Humano A/patogenicidade , Enterovirus Humano A/ultraestrutura , Infecções por Enterovirus/tratamento farmacológico , Infecções por Enterovirus/metabolismo , Infecções por Enterovirus/patologia , Infecções por Enterovirus/virologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Membranas Mitocondriais/efeitos dos fármacos , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/ultraestrutura , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/ultraestrutura , Proibitinas , Proteômica/métodos , Interferência de RNA , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Organismos Livres de Patógenos Específicos , Análise de Sobrevida , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
6.
Elife ; 62017 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-28653620

RESUMO

Positive-strand RNA viruses, the largest genetic class of viruses, include numerous important pathogens such as Zika virus. These viruses replicate their RNA genomes in novel, membrane-bounded mini-organelles, but the organization of viral proteins and RNAs in these compartments has been largely unknown. We used cryo-electron tomography to reveal many previously unrecognized features of Flock house nodavirus (FHV) RNA replication compartments. These spherular invaginations of outer mitochondrial membranes are packed with electron-dense RNA fibrils and their volumes are closely correlated with RNA replication template length. Each spherule's necked aperture is crowned by a striking cupped ring structure containing multifunctional FHV RNA replication protein A. Subtomogram averaging of these crowns revealed twelve-fold symmetry, concentric flanking protrusions, and a central electron density. Many crowns were associated with long cytoplasmic fibrils, likely to be exported progeny RNA. These results provide new mechanistic insights into positive-strand RNA virus replication compartment structure, assembly, function and control.


Assuntos
Microscopia Crioeletrônica , Tomografia com Microscopia Eletrônica , Nodaviridae/fisiologia , RNA Viral/metabolismo , Replicação Viral , Animais , Linhagem Celular , Drosophila , Membranas Mitocondriais/ultraestrutura , Membranas Mitocondriais/virologia
7.
J Virol ; 90(7): 3676-83, 2016 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-26792749

RESUMO

UNLABELLED: Viruses that generate double-stranded RNA (dsRNA) during replication must overcome host defense systems designed to detect this infection intermediate. All positive-sense RNA viruses studied to date modify host membranes to help facilitate the sequestration of dsRNA from host defenses and concentrate replication factors to enhance RNA production. Flock House virus (FHV) is an attractive model for the study of these processes since it is well characterized and infects Drosophila cells, which are known to have a highly effective RNA silencing system. During infection, FHV modifies the outer membrane of host mitochondria to form numerous membrane invaginations, called spherules, that are ∼50 nm in diameter and known to be the site of viral RNA replication. While previous studies have outlined basic structural features of these invaginations, very little is known about the mechanism underlying their formation. Here we describe the optimization of an experimental system for the analysis of FHV host membrane modifications using crude mitochondrial preparations from infected Drosophila cells. These preparations can be programmed to synthesize both single- and double-stranded FHV RNA. The system was used to demonstrate that dsRNA is protected from nuclease digestion by virus-induced membrane invaginations and that spherules play an important role in stimulating RNA replication. Finally, we show that spherules generated during FHV infection appear to be dynamic as evidenced by their ability to form or disperse based on the presence or absence of RNA synthesis. IMPORTANCE: It is well established that positive-sense RNA viruses induce significant membrane rearrangements in infected cells. However, the molecular mechanisms underlying these rearrangements, particularly membrane invagination and spherule formation, remain essentially unknown. How the formation of spherules enhances viral RNA synthesis is also not understood, although it is assumed to be partly a result of evading host defense pathways. To help interrogate some of these issues, we optimized a cell-free replication system consisting of mitochondria isolated from Flock House virus-infected Drosophila cells for use in biochemical and structural studies. Our data suggest that spherules generated during Flock House virus replication are dynamic, protect double-stranded RNA, and enhance RNA replication in general. Cryo-electron microscopy suggests that the samples are amenable to detailed structural analyses of spherules engaged in RNA synthesis. This system thus provides a foundation for understanding the molecular mechanisms underlying spherule formation, maintenance, and function during positive-sense viral RNA replication.


Assuntos
Interações Hospedeiro-Patógeno , Membranas Mitocondriais/virologia , Nodaviridae/fisiologia , Replicação Viral , Animais , Linhagem Celular , Microscopia Crioeletrônica , Drosophila , Membranas Mitocondriais/ultraestrutura , Nodaviridae/ultraestrutura
8.
PLoS Pathog ; 10(10): e1004388, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25329172

RESUMO

Viruses recruit cellular membranes and subvert cellular proteins involved in lipid biosynthesis to build viral replicase complexes and replication organelles. Among the lipids, sterols are important components of membranes, affecting the shape and curvature of membranes. In this paper, the tombusvirus replication protein is shown to co-opt cellular Oxysterol-binding protein related proteins (ORPs), whose deletion in yeast model host leads to decreased tombusvirus replication. In addition, tombusviruses also subvert Scs2p VAP protein to facilitate the formation of membrane contact sites (MCSs), where membranes are juxtaposed, likely channeling lipids to the replication sites. In all, these events result in redistribution and enrichment of sterols at the sites of viral replication in yeast and plant cells. Using in vitro viral replication assay with artificial vesicles, we show stimulation of tombusvirus replication by sterols. Thus, co-opting cellular ORP and VAP proteins to form MCSs serves the virus need to generate abundant sterol-rich membrane surfaces for tombusvirus replication.


Assuntos
Membranas Mitocondriais/virologia , Esteróis/metabolismo , Tombusvirus/fisiologia , Proteínas Virais/metabolismo , Replicação Viral , Replicação do DNA/genética , Membranas Mitocondriais/metabolismo , Saccharomyces cerevisiae
9.
PLoS One ; 9(1): e86124, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24465910

RESUMO

Proper functioning of the mitochondria is crucial for the survival of the cell. Viruses are able to interfere with mitochondrial functions as they infect the host cell. Parvoviruses are known to induce apoptosis in infected cells, but the role of the mitochondria in parvovirus induced cytopathy is only partially known. Here we demonstrate with confocal and electron microscopy that canine parvovirus (CPV) associated with the mitochondrial outer membrane from the onset of infection. During viral entry a transient depolarization of the mitochondrial transmembrane potential and increase in ROS level was detected. Subsequently, mitochondrial homeostasis was normalized shortly, as detected by repolarization of the mitochondrial membrane and decrease of ROS. Indeed, activation of cell survival signalling through ERK1/2 cascade was observed early in CPV infected cells. At 12 hours post infection, concurrent with the expression of viral non-structural protein 1, damage to the mitochondrial structure and depolarization of its membrane were apparent. Results of this study provide additional insight of parvovirus pathology and also more general information of virus-mitochondria association.


Assuntos
Mitocôndrias/metabolismo , Infecções por Parvoviridae/patologia , Infecções por Parvoviridae/virologia , Parvovirus Canino/fisiologia , Animais , Cálcio/metabolismo , Gatos , Linhagem Celular , Cães , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sistema de Sinalização das MAP Quinases , Potencial da Membrana Mitocondrial , Mitocôndrias/patologia , Mitocôndrias/ultraestrutura , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/ultraestrutura , Membranas Mitocondriais/virologia , Espécies Reativas de Oxigênio/metabolismo
10.
PLoS One ; 8(9): e75625, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24086592

RESUMO

Papillomavirus E2 proteins are predominantly retained in the nuclei of infected cells, but oncogenic (high-risk) HPV-18 and 16 E2 can shuttle between the host nucleus and cytoplasm. We show here that cytoplasmic HPV-18 E2 localizes to mitochondrial membranes, and independent mass spectrometry analyses of the E2 interactome revealed association to the inner mitochondrial membrane including components of the respiratory chain. Mitochondrial E2 association modifies the cristae morphology when analyzed by electron microscopy and increases production of mitochondrial ROS. This ROS release does not induce apoptosis, but instead correlates with stabilization of HIF-1α and increased glycolysis. These mitochondrial functions are not shared by the non-oncogenic (low-risk) HPV-6 E2 protein, suggesting that modification of cellular metabolism by high-risk HPV E2 proteins could play a role in carcinogenesis by inducing the Warburg effect.


Assuntos
Papillomavirus Humano 18/metabolismo , Membranas Mitocondriais/metabolismo , Proteínas Oncogênicas Virais/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Virais/metabolismo , Apoptose/fisiologia , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Citoplasma/metabolismo , Citoplasma/virologia , Glicólise/fisiologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/virologia , Membranas Mitocondriais/virologia
11.
Virology ; 439(2): 140-51, 2013 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-23490047

RESUMO

One common feature of positive-strand RNA viruses is the association of viral RNA and viral RNA replicase proteins with specific intracellular membranes to form RNA replication complexes. Wuhan nodavirus (WhNV) encodes protein A, which is the sole viral RNA replicase. Here, we showed that WhNV protein A closely associates with mitochondrial outer membranes and colocalizes with viral RNA replication sites. We further identified the transmembrane domains (N-terminal aa 33-64 and aa 212-254) of protein A for membrane association and mitochondrial localization. Moreover, we found that protein A accumulates genomic RNA by stabilizing the RNA. And our further investigation revealed that the ability of WhNV protein A to associate with membranes is closely linked with its ability for membrane recruitment and stabilization of viral genomic RNA templates. This study represents an advance toward understanding the mechanism of the RNA replication of WhNV and probably other nodaviruses.


Assuntos
Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/virologia , Nodaviridae/fisiologia , RNA Viral/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Replicação Viral , Animais , Nodaviridae/enzimologia , Sinais Direcionadores de Proteínas , Estabilidade de RNA , Proteínas Virais/metabolismo
12.
J Virol ; 86(23): 12779-94, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22973028

RESUMO

Replication of plus-stranded RNA viruses takes place on membranous structures derived from various organelles in infected cells. Previous works with Tomato bushy stunt tombusvirus (TBSV) revealed the recruitment of either peroxisomal or endoplasmic reticulum (ER) membranes for replication. In case of Carnation Italian ringspot tombusvirus (CIRV), the mitochondrial membranes supported CIRV replication. In this study, we developed ER and mitochondrion-based in vitro tombusvirus replication assays. Using purified recombinant TBSV and CIRV replication proteins, we showed that TBSV could use the purified yeast ER and mitochondrial preparations for complete viral RNA replication, while CIRV preferentially replicated in the mitochondrial membranes. The viral RNA became partly RNase resistant after ∼40 to 60 min of incubation in the purified ER and mitochondrial preparations, suggesting that assembly of TBSV and CIRV replicases could take place in the purified ER and mitochondrial membranes in vitro. Using chimeric and heterologous combinations of replication proteins, we showed that multiple domains within the replication proteins are involved in determining the efficiency of tombusvirus replication in the two subcellular membranes. Altogether, we demonstrated that TBSV is less limited while CIRV is more restricted in utilizing various intracellular membranes for replication. Overall, the current work provides evidence that tombusvirus replication could occur in vitro in isolated subcellular membranes, suggesting that tombusviruses have the ability to utilize alternative organellar membranes during infection that could increase the chance of mixed virus replication and rapid evolution during coinfection.


Assuntos
Retículo Endoplasmático/virologia , Membranas Mitocondriais/virologia , Tombusvirus/fisiologia , Replicação Viral/fisiologia , Agrobacterium tumefaciens , Northern Blotting , Western Blotting , Primers do DNA/genética , Técnicas In Vitro , Microscopia Confocal , Reação em Cadeia da Polimerase , Saccharomyces cerevisiae
13.
Virus Res ; 163(2): 580-91, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22222362

RESUMO

Replication of all positive RNA viruses occurs in association with intracellular membranes. In many cases, the mechanism of membrane targeting is unknown and there appears to be no correlation between virus phylogeny and the membrane systems recruited for replication. Pelargonium flower break virus (PFBV, genus Carmovirus, family Tombusviridae) encodes two proteins, p27 and its read-through product p86 (the viral RNA dependent-RNA polymerase), that are essential for replication. Recent reports with other members of the family Tombusviridae have shown that the smaller replicase protein is targeted to specific intracellular membranes and it is assumed to determine the subcellular localization of the replication complex. Using in vivo expression of green fluorescent protein (GFP) fusions in plant and yeast cells, we show here that PFBV p27 localizes in mitochondria. The same localization pattern was found for p86 that contains the p27 sequence at its N-terminus. Cellular fractionation of p27GFP-expressing cells confirmed the confocal microscopy observations and biochemical treatments suggested a tight association of the protein to membranes. Analysis of deletion mutants allowed identification of two regions required for targeting of p27 to mitochondria. These regions mapped toward the N- and C-terminus of the protein, respectively, and could function independently though with distinct efficiency. In an attempt to search for putative cellular factors involved in p27 localization, the subcellular distribution of the protein was checked in a selected series of knockout yeast strains and the outcome of this approach is discussed.


Assuntos
Carmovirus/fisiologia , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/virologia , RNA Polimerase Dependente de RNA/metabolismo , Proteínas Virais/metabolismo , Replicação Viral , Fusão Gênica Artificial , Fracionamento Celular , Células Cultivadas , Análise Mutacional de DNA , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Microscopia Confocal , Sinais Direcionadores de Proteínas , Transporte Proteico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência , Coloração e Rotulagem/métodos , Nicotiana/virologia
14.
J Virol ; 85(5): 2100-11, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21177823

RESUMO

The human cytomegalovirus (HCMV) protein UL37 exon 1 (pUL37x1), also known as viral mitochondrion-localized inhibitor of apoptosis (vMIA), sequentially traffics from the endoplasmic reticulum (ER) through mitochondrion-associated membranes (MAMs) to the outer mitochondrial membrane (OMM), where it robustly inhibits apoptosis. Here, we report the association of pUL37x1/vMIA with internal lipid rafts (LRs) in the ER/MAM. The MAM, which serves as a site for lipid transfer and calcium signaling to mitochondria, is enriched in detergent-resistant membrane (DRM)-forming lipids, including cholesterol and ceramide, which are found in lower concentrations in the bulk ER. Sigma 1 receptor (Sig-1R), a MAM chaperone affecting calcium signaling to mitochondria, is anchored in the MAM by its LR association. Because of its trafficking through the MAM and partial colocalization with Sig-1R, we tested whether pUL37x1/vMIA associates with MAM LRs. Extraction with methyl-ß-cyclodextrin (MßCD) removed pUL37x1/vMIA from lysed but not intact cells, indicating its association with internal LRs. Furthermore, the isolation of DRMs from purified intracellular organelles independently verified the localization of pUL37x1/vMIA within ER/MAM LRs. However, pUL37x1/vMIA was not detected in DRMs from mitochondria. pUL37x1/vMIA associated with LRs during all temporal phases of HCMV infection, indicating the likely importance of this location for HCMV growth. Although detected during its sequential trafficking to the OMM, the pUL37x1/vMIA LR association was independent of its mitochondrial targeting signals. Rather, it was dependent upon cholesterol binding. These studies suggest a conserved ability of UL37 proteins to interact with cholesterol and LRs, which is functionally distinguishable from their sequential trafficking to mitochondria.


Assuntos
Infecções por Citomegalovirus/metabolismo , Citomegalovirus/metabolismo , Éxons , Proteínas Imediatamente Precoces/metabolismo , Lipídeos de Membrana/metabolismo , Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/virologia , Células HeLa , Humanos , Proteínas Imediatamente Precoces/genética , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/virologia
15.
Rev Med Virol ; 19(3): 147-64, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19367604

RESUMO

By exploiting host cell machineries, viruses provide powerful tools for gaining insight into cellular pathways. Proteins from two unrelated viruses, human CMV (HCMV) and HCV, are documented to traffic sequentially from the ER into mitochondria, probably through the mitochondria-associated membrane (MAM) compartment. The MAM are sites of ER-mitochondrial contact enabling the direct transfer of membrane bound lipids and the generation of high calcium (Ca2+) microdomains for mitochondria signalling and responses to cellular stress. Both HCV core protein and HCMV UL37 proteins are associated with Ca2+ regulation and apoptotic signals. Trafficking of viral proteins to the MAM may allow viruses to manipulate a variety of fundamental cellular processes, which converge at the MAM, including Ca2+ signalling, lipid synthesis and transfer, bioenergetics, metabolic flow, and apoptosis. Because of their distinct topologies and targeted MAM sub-domains, mitochondrial trafficking (albeit it through the MAM) of the HCMV and HCV proteins predictably involves alternative pathways and, hence, distinct targeting signals. Indeed, we found that multiple cellular and viral proteins, which target the MAM, showed no apparent consensus primary targeting sequences. Nonetheless, these viral proteins provide us with valuable tools to access the poorly characterised MAM compartment, to define its cellular constituents and describe how virus infection alters these to its own end. Furthermore, because proper trafficking of viral proteins is necessary for their function, discovering the requirements for MAM to mitochondrial trafficking of essential viral proteins may provide novel targets for the rational design of anti-viral drugs.


Assuntos
Infecções por Citomegalovirus/virologia , Citomegalovirus/metabolismo , Hepacivirus/metabolismo , Hepatite C/virologia , Membranas Mitocondriais/metabolismo , Proteínas Virais/metabolismo , Animais , Sinalização do Cálcio , Infecções por Citomegalovirus/metabolismo , Hepatite C/metabolismo , Humanos , Membranas Mitocondriais/virologia , Transporte Proteico
16.
Virology ; 385(2): 444-54, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19136133

RESUMO

The functions of the Betanodavirus non-structural protein B1 is still unknown. We examined B1 expression patterns and investigated novel cell death regulatory functions for this viral protein following RGNNV infection in fish cells. The B1 gene (336 nt) was cloned from the redspotted grouper nervous necrosis virus (RGNNV) genome. B1 mRNA was rapidly expressed in the fish cells from viral RNA3 at 12 h post-infection (p.i.). At the protein level, expression was low at 12 h p.i., and then increased rapidly between 24 h and 72 h p.i. In RGNNV-infected, B1-containing fish cells, over expression of RGNNV B1 reduced Annexin-V positive cells by 50% and 65% at 48 h and 72 h p.i., respectively, and decreased loss of mitochondrial membrane potential (MMP) by 20% and 70% at 48 h and 72 h p.i., respectively. Finally, B1 knockdown during RGNNV infection using anti-sense RNA increased necrotic cell death and reduced cell viability during the early replication cycle (24 h p.i.). Our results suggest that B1 is an early expression protein that has an anti-necrotic cell death function which reduces the MMP loss and enhances viral host cell viability. This finding provides new insights into RNA viral pathogenesis and disease control.


Assuntos
Peixes/virologia , Nodaviridae/genética , Nodaviridae/metabolismo , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Morte Celular/fisiologia , Linhagem Celular , Clonagem Molecular , Regulação Viral da Expressão Gênica , Técnicas de Silenciamento de Genes , Membranas Mitocondriais/virologia , Dados de Sequência Molecular , RNA Viral/genética , RNA Viral/metabolismo , Alinhamento de Sequência , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética
17.
PLoS Biol ; 5(9): e220, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17696647

RESUMO

Positive-strand RNA viruses are the largest genetic class of viruses and include many serious human pathogens. All positive-strand RNA viruses replicate their genomes in association with intracellular membrane rearrangements such as single- or double-membrane vesicles. However, the exact sites of RNA synthesis and crucial topological relationships between relevant membranes, vesicle interiors, surrounding lumens, and cytoplasm generally are poorly defined. We applied electron microscope tomography and complementary approaches to flock house virus (FHV)-infected Drosophila cells to provide the first 3-D analysis of such replication complexes. The sole FHV RNA replication factor, protein A, and FHV-specific 5-bromouridine 5'-triphosphate incorporation localized between inner and outer mitochondrial membranes inside approximately 50-nm vesicles (spherules), which thus are FHV-induced compartments for viral RNA synthesis. All such FHV spherules were outer mitochondrial membrane invaginations with interiors connected to the cytoplasm by a necked channel of approximately 10-nm diameter, which is sufficient for ribonucleotide import and product RNA export. Tomographic, biochemical, and other results imply that FHV spherules contain, on average, three RNA replication intermediates and an interior shell of approximately 100 membrane-spanning, self-interacting protein As. The results identify spherules as the site of protein A and nascent RNA accumulation and define spherule topology, dimensions, and stoichiometry to reveal the nature and many details of the organization and function of the FHV RNA replication complex. The resulting insights appear relevant to many other positive-strand RNA viruses and support recently proposed structural and likely evolutionary parallels with retrovirus and double-stranded RNA virus virions.


Assuntos
Membranas Mitocondriais/virologia , Organelas/virologia , RNA Viral/biossíntese , Replicação Viral , Animais , Drosophila/citologia , Drosophila/virologia , Imageamento Tridimensional , Microscopia Eletrônica , Nodaviridae
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...