Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
J Virol ; 96(6): e0175721, 2022 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-35107373

RESUMO

Emerging viruses impose global threats to animal and human populations and may bear novel genes with limited homology to known sequences, necessitating the development of novel approaches to infer and test protein functions. This challenge is dramatically evident in tilapia lake virus (TiLV), an emerging "orthomyxo-like" virus that threatens the global tilapia aquaculture and food security of millions of people. The majority of TiLV proteins have no homology to known sequences, impeding functionality assessments. Using a novel bioinformatics approach, we predicted that TiLV's Protein 4 encodes the nucleoprotein, a factor essential for viral RNA replication. Multiple methodologies revealed the expected properties of orthomyxoviral nucleoproteins. A modified yeast three-hybrid assay detected Protein 4-RNA interactions, which were independent of the RNA sequence, and identified specific positively charged residues involved. Protein 4-RNA interactions were uncovered by R-DeeP and XRNAX methodologies. Immunoelectron microscopy found that multiple Protein 4 copies localized along enriched ribonucleoproteins. TiLV RNA from cells and virions coimmunoprecipitated with Protein 4. Immunofluorescence microscopy detected Protein 4 in the cytoplasm and nuclei, and nuclear Protein 4 increased upon CRM1 inhibition, suggesting CRM1-dependent nuclear export of TiLV RNA. Together, these data reveal TiLV's nucleoprotein and highlight the ability to infer protein functionality, including novel RNA-binding proteins, in emerging pathogens. These are important in light of the expected discovery of many unknown viruses and the zoonotic potential of such pathogens. IMPORTANCE Tilapia is an important source of dietary protein, especially in developing countries. Massive losses of tilapia were identified worldwide, risking the food security of millions of people. Tilapia lake virus (TiLV) is an emerging pathogen responsible for these disease outbreaks. TiLV's genome encodes 10 major proteins, 9 of which show no homology to other known viral or cellular proteins, hindering functionality assessment of these proteins. Here, we describe a novel bioinformatics approach to infer the functionality of TiLV proteins, which predicted Protein 4 as the nucleoprotein, a factor essential for viral RNA replication. We provided experimental support for this prediction by applying multiple molecular, biochemical, and imaging approaches. Overall, we illustrate a strategy for functional analyses in viral discovery. The strategy is important in light of the expected discovery of many unknown viruses and the zoonotic potential of such pathogens.


Assuntos
Nucleoproteínas , Vírus de RNA , Tilápia , Animais , Doenças dos Peixes/virologia , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , Infecções por Vírus de RNA/virologia , Vírus de RNA/classificação , Vírus de RNA/genética , Vírus de RNA/patogenicidade , RNA Viral/genética , Tilápia/genética
2.
Nat Methods ; 16(4): 327-332, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30886410

RESUMO

Single-cell RNA sequencing (scRNA-seq) is a rich resource of cellular heterogeneity, opening new avenues in the study of complex tissues. We introduce Cell Population Mapping (CPM), a deconvolution algorithm in which reference scRNA-seq profiles are leveraged to infer the composition of cell types and states from bulk transcriptome data ('scBio' CRAN R-package). Analysis of individual variations in lungs of influenza-virus-infected mice reveals that the relationship between cell abundance and clinical symptoms is a cell-state-specific property that varies gradually along the continuum of cell-activation states. The gradual change is confirmed in subsequent experiments and is further explained by a mathematical model in which clinical outcomes relate to cell-state dynamics along the activation process. Our results demonstrate the power of CPM in reconstructing the continuous spectrum of cell states within heterogeneous tissues.


Assuntos
Biologia Computacional , Genômica , Análise de Sequência de RNA , Análise de Célula Única , Algoritmos , Animais , Separação Celular , Feminino , Fibroblastos/metabolismo , Citometria de Fluxo , Perfilação da Expressão Gênica , Genoma Humano , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Pulmão/virologia , Cadeias de Markov , Camundongos , Camundongos Endogâmicos C57BL , Orthomyxoviridae , Fagócitos/metabolismo , Valores de Referência , Software , Transcriptoma
3.
Int J Cancer ; 148(9): 2321-2334, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33197301

RESUMO

STAT1 is a critical effector and a target gene of interferon (IFN) signaling, and thus a central mediator of antiviral responses. As both a mediator and a target of IFN signals, STAT1 expression reports on, and determines IFN activity. Gene expression analyses of melanoma patient samples revealed varied levels of STAT1 expression, which highly correlated with expression of >700 genes. The ability of oncolytic viruses to exploit tumor-induced defects to antiviral responses suggests that oncolytic viruses may efficiently target a subset of melanomas, yet these should be defined. We modeled this scenario with murine B16F10 melanomas, immortalized skin fibroblasts as controls and a novel oncolytic virus, EHDV-TAU. In B16F10 cells, constitutive low expression of STAT1 and its target genes, which included intracellular pattern recognition receptors (PRRs), correlated with their inability to mount IFN-based antiviral responses upon EHDV-TAU challenge, and with potency of EHDV-TAU-induced oncolysis. This underexpression of interferon stimulated genes (ISGs) and PRRs, and the inability of EHDV-TAU to induce their expression, were reversed by epigenetic modifiers, suggesting epigenetic silencing as a basis for their underexpression. Despite their inability to mount IFN/STAT-based responses upon viral infection, EHDV-TAU infected B16F10 cells secreted immune-stimulatory chemokines. Accordingly, in vivo, EHDV-TAU enhanced intratumoral infiltration of cytotoxic T-cells and reduced growth of local and distant tumors. We propose that "STAT1 signatures" should guide melanoma virotherapy treatments, and that oncolytic viruses such as EHDV-TAU have the potential to exploit the cellular context of low-STAT1 tumors.


Assuntos
Antivirais/uso terapêutico , Melanoma/tratamento farmacológico , Vírus Oncolíticos/patogenicidade , Animais , Antivirais/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Camundongos
4.
Eur J Immunol ; 47(4): 692-703, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28191644

RESUMO

Natural killer (NK) cells are capable of killing various pathogens upon stimulation of activating receptors. Human metapneumovirus (HMPV) is a respiratory virus, which was discovered in 2001 and is responsible for acute respiratory tract infection in infants and children worldwide. HMPV infection is very common, infecting around 70% of all children under the age of five. Under immune suppressive conditions, HMPV infection can be fatal. Not much is known on how NK cells respond to HMPV. In this study, using reporter assays and NK-cell cytotoxicity assays performed with human and mouse NK cells, we demonstrated that the NKp46-activating receptor and its mouse orthologue Ncr1, both members of the natural cytotoxicity receptor (NCR) family, recognized an unknown ligand expressed by HMPV-infected human cells. We demonstrated that MHC class I is upregulated and MICA is downregulated upon HMPV infection. We also characterized mouse NK-cell phenotype in the blood and the lungs of HMPV-infected mice and found that lung NK cells are more activated and expressing NKG2D, CD43, CD27, KLRG1, and CD69 compared to blood NK cells regardless of HMPV infection. Finally, we demonstrated, using Ncr1-deficient mice, that NCR1 plays a critical role in controlling HMPV infection.


Assuntos
Antígenos Ly/metabolismo , Células Matadoras Naturais/imunologia , Pulmão/imunologia , Metapneumovirus/imunologia , Receptor 1 Desencadeador da Citotoxicidade Natural/metabolismo , Infecções por Paramyxoviridae/imunologia , Animais , Antígenos Ly/genética , Criança , Citotoxicidade Imunológica , Células HEK293 , Humanos , Lactente , Células Matadoras Naturais/virologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptor 1 Desencadeador da Citotoxicidade Natural/genética , Carga Viral
5.
J Clin Microbiol ; 55(3): 759-767, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27974544

RESUMO

Tilapia are an important group of farmed fish that serve as a significant protein source worldwide. In recent years, substantial mortality of wild tilapia has been observed in the Sea of Galilee and in commercial ponds in Israel and Ecuador. We have identified the etiological agent of these mass die-offs as a novel orthomyxo-like virus and named it tilapia lake virus (TiLV). Here, we provide the conditions for efficient isolation, culturing, and quantification of the virus, including the use of susceptible fish cell lines. Moreover, we describe a sensitive nested reverse transcription-PCR (RT-PCR) assay allowing the rapid detection of TiLV in fish organs. This assay revealed, for the first time to our knowledge, the presence of TiLV in diseased Colombian tilapia, indicating a wider distribution of this emerging pathogen and stressing the risk that TiLV poses for the global tilapia industry. Overall, the described procedures should provide the tilapia aquaculture industry with important tools for the detection and containment of this pathogen.


Assuntos
Doenças dos Peixes/diagnóstico , Infecções por Orthomyxoviridae/veterinária , Orthomyxoviridae/isolamento & purificação , RNA Viral/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Tilápia/virologia , Cultura de Vírus/métodos , Animais , Linhagem Celular , Colômbia , Doenças dos Peixes/virologia , Infecções por Orthomyxoviridae/diagnóstico , Infecções por Orthomyxoviridae/virologia , Reação em Cadeia da Polimerase/métodos , RNA Viral/genética
6.
Mol Cell Proteomics ; 14(3): 532-43, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25556234

RESUMO

Although human respiratory syncytial virus (RSV) is the most common cause of bronchiolitis and pneumonia in infants and elderly worldwide, there is no licensed RSV vaccine or effective drug treatment available. The RSV Matrix protein plays key roles in virus life cycle, being found in the nucleus early in infection in a transcriptional inhibitory role, and later localizing in viral inclusion bodies before coordinating viral assembly and budding at the plasma membrane. In this study, we used a novel, high throughput microfluidics platform and custom human open reading frame library to identify novel host cell binding partners of RSV matrix. Novel interactors identified included proteins involved in host transcription regulation, the innate immunity response, cytoskeletal regulation, membrane remodeling, and cellular trafficking. A number of these interactions were confirmed by immunoprecipitation and cellular colocalization approaches. Importantly, the physiological significance of matrix interaction with the actin-binding protein cofilin 1, caveolae protein Caveolin 2, and the zinc finger protein ZNF502 was confirmed. siRNA knockdown of the host protein levels resulted in reduced RSV virus production in infected cells. These results have important implications for future antiviral strategies aimed at targets of RSV matrix in the host cell.


Assuntos
Proteínas de Transporte/metabolismo , Caveolina 2/metabolismo , Cofilina 1/metabolismo , Técnicas Analíticas Microfluídicas/métodos , Proteínas Nucleares/metabolismo , Vírus Sinciciais Respiratórios/fisiologia , Proteínas da Matriz Viral/metabolismo , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Chlorocebus aethiops , Biblioteca Gênica , Células HEK293 , Humanos , Fases de Leitura Aberta , Células Vero , Replicação Viral
7.
PLoS Pathog ; 10(10): e1004474, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25356837

RESUMO

The murine leukaemia virus (MLV) gag gene encodes a small protein called p12 that is essential for the early steps of viral replication. The N- and C-terminal regions of p12 are sequentially acting domains, both required for p12 function. Defects in the C-terminal domain can be overcome by introducing a chromatin binding motif into the protein. However, the function of the N-terminal domain remains unknown. Here, we undertook a detailed analysis of the effects of p12 mutation on incoming viral cores. We found that both reverse transcription complexes and isolated mature cores from N-terminal p12 mutants have altered capsid complexes compared to wild type virions. Electron microscopy revealed that mature N-terminal p12 mutant cores have different morphologies, although immature cores appear normal. Moreover, in immunofluorescent studies, both p12 and capsid proteins were lost rapidly from N-terminal p12 mutant viral cores after entry into target cells. Importantly, we determined that p12 binds directly to the MLV capsid lattice. However, we could not detect binding of an N-terminally altered p12 to capsid. Altogether, our data imply that p12 stabilises the mature MLV core, preventing premature loss of capsid, and that this is mediated by direct binding of p12 to the capsid shell. In this manner, p12 is also retained in the pre-integration complex where it facilitates tethering to mitotic chromosomes. These data also explain our previous observations that modifications to the N-terminus of p12 alter the ability of particles to abrogate restriction by TRIM5alpha and Fv1, factors that recognise viral capsid lattices.


Assuntos
Capsídeo/metabolismo , Produtos do Gene gag/metabolismo , Vírus da Leucemia Murina/genética , Infecções por Retroviridae/virologia , Replicação Viral , Sequência de Aminoácidos , Animais , Capsídeo/ultraestrutura , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Cromossomos , Produtos do Gene gag/genética , Humanos , Vírus da Leucemia Murina/fisiologia , Vírus da Leucemia Murina/ultraestrutura , Camundongos , Dados de Sequência Molecular , Mutação , Estrutura Terciária de Proteína , Proteínas Recombinantes , Transcrição Reversa , Alinhamento de Sequência , Vírion
8.
Retrovirology ; 12: 95, 2015 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-26577111

RESUMO

BACKGROUND: Murine leukemia viruses (MLVs) naturally infect unsynchronized T and B lymphocytes, thus, the incoming virus encounters both interphase and mitotic cells. While it is well accepted that MLV requires cell division to complete its replication cycle, it is not known if ab initio infection of mitotic cells can result in productive infection. This question is highly relevant since the milieu of mitotic cells is markedly different from this of interphase cells; e.g. lacking radial microtubule network and intact nuclear envelope. To follow MLV infection in mitotic and interphase cells in real-time, we employed our recently developed infectious MLV particles with labeled cores, cellular models expressing fluorescence markers of different intracellular compartments and protocols for reversible mitotic arrest of MLV-susceptible cells. RESULTS: Multi-wavelength live cell imaging was employed to simultaneously visualize GFP-labeled MLV cores, DiD-labeled viral or cellular membranes, and fluorescently-labeled microtubules or chromosomes. Cells were imaged either at interphase or upon mitotic arrest with microtubule poisons. Analysis of virus localization and trajectories revealed entry by endocytosis at interphase and mitosis, and correlation between viral mobility parameters and presence or absence of polymerized interphase microtubules. The success of infection of viruses that entered cells in mitosis was evidenced by their ability to reverse transcribe, their targeting to condensed chromosomes in the absence of radial microtubule network, and gene expression upon exit from mitosis. Comparison of infection by N, B or NB -tropic viruses in interphase and mitotic human cells revealed reduced restriction of the N-tropic virus, for infection initiated in mitosis. CONCLUSIONS: The milieu of the mitotic cells supports all necessary requirements for early stages of MLV infection. Such milieu is suboptimal for restriction of N-tropic viruses, most likely by TRIM5α.


Assuntos
Interfase , Vírus da Leucemia Murina/genética , Vírus da Leucemia Murina/fisiologia , Mitose , Vírion/fisiologia , Animais , Fatores de Restrição Antivirais , Proteínas de Transporte/metabolismo , Linhagem Celular , Cromossomos/virologia , Interações Hospedeiro-Patógeno , Humanos , Vírus da Leucemia Murina/ultraestrutura , Camundongos , Células NIH 3T3 , Membrana Nuclear/virologia , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases , Vírion/ultraestrutura , Integração Viral , Replicação Viral
9.
J Clin Microbiol ; 52(12): 4137-46, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25232154

RESUMO

Tilapines are important for the sustainability of ecological systems and serve as the second most important group of farmed fish worldwide. Significant mortality of wild and cultured tilapia has been observed recently in Israel. The etiological agent of this disease, a novel RNA virus, is described here, and procedures allowing its isolation and detection are revealed. The virus, denominated tilapia lake virus (TiLV), was propagated in primary tilapia brain cells or in an E-11 cell line, and it induced a cytopathic effect at 5 to 10 days postinfection. Electron microscopy revealed enveloped icosahedral particles of 55 to 75 nm. Low-passage TiLV, injected intraperitoneally in tilapia, induced a disease resembling the natural disease, which typically presents with lethargy, ocular alterations, and skin erosions, with >80% mortality. Histological changes included congestion of the internal organs (kidneys and brain) with foci of gliosis and perivascular cuffing of lymphocytes in the brain cortex; ocular inflammation included endophthalmitis and cataractous changes of the lens. The cohabitation of healthy and diseased fish demonstrated that the disease is contagious and that mortalities (80 to 100%) occur within a few days. Fish surviving the initial mortality were immune to further TiLV infections, suggesting the mounting of a protective immune response. Screening cDNA libraries identified a TiLV-specific sequence, allowing the design of a PCR-based diagnostic test. This test enables the specific identification of TiLV in tilapines and should help control the spread of this virus worldwide.


Assuntos
Infecções por Vírus de RNA/veterinária , Vírus de RNA/classificação , Vírus de RNA/isolamento & purificação , Tilápia/virologia , Animais , Encéfalo/patologia , Células Cultivadas , Efeito Citopatogênico Viral , Olho/patologia , Fibroblastos/virologia , Israel , Rim/patologia , Microscopia Eletrônica de Transmissão , Dados de Sequência Molecular , Infecções por Vírus de RNA/patologia , Infecções por Vírus de RNA/transmissão , Infecções por Vírus de RNA/virologia , Vírus de RNA/genética , RNA Viral/genética , Análise de Sequência de DNA , Análise de Sobrevida , Vírion/ultraestrutura , Cultura de Vírus
10.
J Virol ; 87(24): 13397-408, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24089565

RESUMO

The mode and timing of virally induced cell death hold the potential of regulating viral yield, viral transmission, and the severity of virally induced disease. Orbiviruses such as the epizootic hemorrhagic disease virus (EHDV) are nonenveloped and cytolytic. To date, the death of cells infected with EHDV, the signal transduction pathways involved in this process, and the consequence of their inhibition have yet to be characterized. Here, we report that the Ibaraki strain of EHDV2 (EHDV2-IBA) induces apoptosis, autophagy, a decrease in cellular protein synthesis, the activation of c-Jun N-terminal kinase (JNK), and the phosphorylation of the JNK substrate c-Jun. The production of infectious virions decreased upon inhibition of apoptosis with the pan-caspase inhibitor Q-VD-OPH (quinolyl-valyl-O-methylaspartyl-[-2,6-difluorophenoxy]-methyl ketone), upon inhibition of autophagy with 3-methyladenine or via the knockout of the autophagy regulator Atg5, or upon treatment of infected cells with the JNK inhibitor SP600125 or the cyclin-dependent kinase (CDK) inhibitor roscovitine, which also inhibited c-Jun phosphorylation. Moreover, Q-VD-OPH, SP600125, and roscovitine partially reduced EHDV2-IBA-induced cell death, and roscovitine diminished the induction of autophagy by EHDV2-IBA. Taken together, our results imply that EHDV induces and benefits from the activation of signaling pathways involved in cell stress and death.


Assuntos
Apoptose , Autofagia , Doenças dos Bovinos/fisiopatologia , Vírus da Doença Hemorrágica Epizoótica/fisiologia , Infecções por Reoviridae/veterinária , Doenças dos Ovinos/fisiopatologia , Animais , Bovinos , Doenças dos Bovinos/genética , Doenças dos Bovinos/metabolismo , Doenças dos Bovinos/virologia , Linhagem Celular , Vírus da Doença Hemorrágica Epizoótica/genética , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Camundongos , Biossíntese de Proteínas , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Infecções por Reoviridae/metabolismo , Infecções por Reoviridae/fisiopatologia , Infecções por Reoviridae/virologia , Ovinos , Doenças dos Ovinos/genética , Doenças dos Ovinos/metabolismo , Doenças dos Ovinos/virologia , Transdução de Sinais
11.
PLoS Pathog ; 8(12): e1003103, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23300449

RESUMO

The p12 protein of the murine leukemia virus (MLV) is a constituent of the pre-integration complex (PIC) but its function in this complex remains unknown. We developed an imaging system to monitor MLV PIC trafficking in live cells. This allowed the visualization of PIC docking to mitotic chromosomes and its release upon exit from mitosis. Docking occurred concomitantly with nuclear envelope breakdown and was impaired for PICs of viruses with lethal p12 mutations. Insertion of a heterologous chromatin binding module into p12 of one of these mutants restored PICs attachment to the chromosomes and partially rescued virus replication. Capsid dissociated from wild type PICs in mitotic cells but remained associated with PICs harboring tethering-negative p12 mutants. Altogether, these results explain, in part, MLV restriction to dividing cells and reveal a role for p12 as a factor that tethers MLV PIC to mitotic chromosomes.


Assuntos
Cromossomos/virologia , Produtos do Gene gag/genética , Produtos do Gene gag/metabolismo , Vírus da Leucemia Murina/genética , Células 3T3 , Animais , Capsídeo , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Cromatina/metabolismo , Camundongos , Mitose , Mutação , Membrana Nuclear/patologia , Membrana Nuclear/virologia , Ligação Proteica , Ligação Viral , Integração Viral , Replicação Viral/genética
12.
mSystems ; 9(4): e0104823, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38446104

RESUMO

Secondary bacterial challenges during influenza virus infection "superinfection") cause excessive mortality and hospitalization. Here, we present a longitudinal study of bulk gene expression changes in murine lungs during superinfection, with an initial influenza A virus infection and a subsequent Streptococcus pneumoniae infection. In addition to the well-characterized impairment of the host response, we identified superinfection-specific alterations in the global transcriptional program that are linked to the host's ability to resist the pathogens. Particularly, whereas superinfected mice manifested an excessive rapid induction of the resistance-to-infection program, there was a substantial tissue-level rewiring of this program: upon superinfection, interferon-regulated genes were switched from positive to negative correlations with the host's resistance state, whereas genes of fatty acid metabolism switched from negative to positive correlations with resistance states. Thus, the transcriptional resistance state in superinfection is reprogrammed toward repressed interferon signaling and induced fatty acid metabolism. Our findings suggest new insights into a tissue-level remodeling of the host defense upon superinfection, providing promising targets for future therapeutic interventions. IMPORTANCE: Secondary bacterial infections are the most frequent complications during influenza A virus (IAV) pandemic outbreaks, contributing to excessive morbidity and mortality in the human population. Most IAV-related deaths are attributed to Streptococcus pneumoniae (SP) infections, which usually begin within the first week of IAV infection in the respiratory tracts. Here, we focused on longitudinal transcriptional responses during a superinfection model consisting of an SP infection that follows an initial IAV infection, comparing superinfection to an IAV-only infection, an SP-only infection, and control treatments. Our longitudinal data allowed a fine analysis of gene expression changes during superinfection. For instance, we found that superinfected mice exhibited rapid gene expression induction or reduction within the first 12 h after encountering the second pathogen. Cell proliferation and immune response activation processes were upregulated, while endothelial processes, vasculogenesis, and angiogenesis were downregulated, providing promising targets for future therapeutic interventions. We further analyzed the longitudinal transcriptional responses in the context of a previously defined spectrum of the host's resistance state, revealing superinfection-specific reprogramming of resistance states, such as reprogramming of fatty acid metabolism and interferon signaling. The reprogrammed functions are compelling new targets for switching the pathogenic superinfection state into a single-infection state.


Assuntos
Vírus da Influenza A , Influenza Humana , Infecções Pneumocócicas , Superinfecção , Camundongos , Humanos , Animais , Streptococcus pneumoniae , Superinfecção/complicações , Estudos Longitudinais , Influenza Humana/genética , Infecções Pneumocócicas/genética , Imunidade Inata/genética , Interferons , Ácidos Graxos
13.
BMC Evol Biol ; 13: 164, 2013 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-23914950

RESUMO

BACKGROUND: Synonymous or silent mutations are usually thought to evolve neutrally. However, accumulating recent evidence has demonstrated that silent mutations may destabilize RNA structures or disrupt cis regulatory motifs superimposed on coding sequences. Such observations suggest the existence of stretches of codon sites that are evolutionary conserved at both DNA-RNA and protein levels. Such stretches may point to functionally important regions within protein coding sequences not necessarily reflecting functional constraints on the amino-acid sequence. The HIV-1 genome is highly compact, and often harbors overlapping functional elements at the protein, RNA, and DNA levels. This superimposition of functions leads to complex selective forces acting on all levels of the genome and proteome. Considering the constraints on HIV-1 to maintain such a highly compact genome, we hypothesized that stretches of synonymous conservation would be common within its genome. RESULTS: We used a combined computational-experimental approach to detect and characterize regions exhibiting strong purifying selection against synonymous substitutions along the HIV-1 genome. Our methodology is based on advanced probabilistic evolutionary models that explicitly account for synonymous rate variation among sites and rate dependencies among adjacent sites. These models are combined with a randomization procedure to automatically identify the most statistically significant regions of conserved synonymous sites along the genome. Using this procedure we identified 21 conserved regions. Twelve of these are mapped to regions within overlapping genes, seven correlate with known functional elements, while the functions of the remaining four are yet unknown. Among these four regions, we chose the one that deviates most from synonymous rate homogeneity for in-depth computational and experimental characterization. In our assays aiming to quantify viral fitness in both early and late stages of the replication cycle, no differences were observed between the mutated and the wild type virus following the introduction of synonymous mutations. CONCLUSIONS: The contradiction between the inferred purifying selective forces and the lack of effect of these mutations on viral replication may be explained by the fact that the phenotype was measured in single-cycle infection assays in cell culture. Such a system does not account for the complexity of HIV-1 infections in vivo, which involves multiple infection cycles and interaction with the host immune system.


Assuntos
Evolução Molecular , Genoma Viral , HIV-1/genética , Proteínas Virais/genética , Sequência de Aminoácidos , Sequência de Bases , Códon/genética , Sequência Conservada , Humanos , Fases de Leitura Aberta
14.
Artigo em Inglês | MEDLINE | ID: mdl-23722834

RESUMO

The Gag precursor is the major structural protein of the virion of human immunodeficiency virus-1 (HIV-1). Capsid protein (CA), a cleavage product of Gag, plays an essential role in virus assembly both in Gag-precursor multimerization and in capsid core formation. The carboxy-terminal domain (CTD) of CA contains 20 residues that are highly conserved across retroviruses and constitute the major homology region (MHR). Genetic evidence implies a role for the MHR in interactions between Gag precursors during the assembly of the virus, but the structural basis for this role remains elusive. This paper describes a novel triclinic structure of the HIV-1 CA CTD at 1.6 Å resolution with two canonical dimers of CA CTD in the asymmetric unit. The canonical dimers form a newly identified packing interface where interactions of four conserved MHR residues take place. This is the first structural indication that these MHR residues participate in the putative CTD-CTD interactions. These findings suggest that the molecules forming this novel interface resemble an intermediate structure that participates in the early steps of HIV-1 assembly. This interface may therefore provide a novel target for antiviral drugs.


Assuntos
Proteínas do Capsídeo/química , HIV-1/química , Multimerização Proteica , Montagem de Vírus/fisiologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Proteínas do Capsídeo/fisiologia , Cristalização , HIV-1/fisiologia , Humanos , Dados de Sequência Molecular , Estrutura Terciária de Proteína
15.
Eur J Cell Biol ; 102(2): 151328, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37321037

RESUMO

Lipocalin-2 (LCN2) performs pleiotropic and tumor context-dependent functions in cancers of diverse etiologies. In prostate cancer (PCa) cells, LCN2 regulates distinct phenotypic features, including cytoskeleton organization and expression of inflammation mediators. Oncolytic virotherapy uses oncolytic viruses (OVs) to kill cancer cells and induce anti-tumor immunity. A main source of specificity of OVs towards tumor cells stems from cancer-induced defects in interferon (IFN)-based cell autonomous immune responses. However, the molecular underpinnings of such defects in PCa cells are only partially understood. Moreover, LCN2 effects on IFN responses of PCa cells and their susceptibility to OVs are unknown. To examine these issues, we queried gene expression databases for genes coexpressed with LCN2, revealing co-expression of IFN-stimulated genes (ISGs) and LCN2. Analysis of human PCa cells revealed correlated expression of LCN2 and subsets of IFNs and ISGs. CRISPR/Cas9-mediated stable knockout of LCN2 in PC3 cells or transient overexpression of LCN2 in LNCaP cells revealed LCN2-mediated regulation of IFNE (and IFNL1) expression, activation of JAK/STAT pathway, and expression of selected ISGs. Accordingly, and dependent on a functional JAK/STAT pathway, LCN2 reduced the susceptibility of PCa cells to infection with the IFN-sensitive OV, EHDV-TAU. In PC3 cells, LCN2 knockout increased phosphorylation of eukaryotic initiation factor 2α (p-eIF2α). Inhibition of PKR-like ER kinase (PERK) in PC3-LCN2-KO cells reduced p-eIF2α while increasing constitutive IFNE expression, phosphorylation of STAT1, and ISG expression; and decreasing EHDV-TAU infection. Together, these data propose that LCN2 regulates PCa susceptibility to OVs through attenuation of PERK activity and increased IFN and ISG expression.


Assuntos
Vírus Oncolíticos , Neoplasias da Próstata , Viroses , Humanos , Masculino , Interferons/genética , Interferons/metabolismo , Janus Quinases/metabolismo , Lipocalina-2/genética , Lipocalina-2/metabolismo , Vírus Oncolíticos/genética , Vírus Oncolíticos/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/terapia , Neoplasias da Próstata/patologia , Transdução de Sinais/fisiologia , Fatores de Transcrição STAT/metabolismo
16.
iScience ; 26(4): 106370, 2023 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-37009225

RESUMO

Rainbow trout (Oncorhynchus mykiss) is the principal species of inland-farmed fish in the Western hemisphere. Recently, we diagnosed in farmed rainbow trout a disease in which the hallmark is granulomatous-like hepatitis. No biotic agents could be isolated from lesions. Still, unbiased high-throughput sequencing and bioinformatics analyses revealed the presence of a novel piscine nidovirus that we named "Trout Granulomatous Virus" (TGV). TGV genome (28,767 nucleotides long) is predicted to encode non-structural (1a and 1 ab) and structural (S, M, and N) proteins that resemble proteins of other known piscine nidoviruses. High loads of TGV transcripts were detected by quantitative RT-PCR in diseased fish and visualized in hepatic granulomatous sites by fluorescence in situ hybridization. Transmission electron microscopy (TEM) revealed coronavirus-like particles in these lesions. Together, these analyses corroborated the association of TGV with the lesions. The identification and detection of TGV provide means to control TGV spread in trout populations.

17.
Retrovirology ; 9: 7, 2012 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-22260459

RESUMO

BACKGROUND: The cellular activity of many factors and pathways is required to execute the complex replication cycle of the human immunodeficiency virus type 1 (HIV-1). To reveal these cellular components, several extensive RNAi screens have been performed, listing numerous 'HIV-dependency factors'. However, only a small overlap between these lists exists, calling for further evaluation of the relevance of specific factors to HIV-1 replication and for the identification of additional cellular candidates. TBC1D20, the GTPase-activating protein (GAP) of Rab1, regulates endoplasmic reticulum (ER) to Golgi trafficking, was not identified in any of these screens, and its involvement in HIV-1 replication cycle is tested here. FINDINGS: Excessive TBC1D20 activity perturbs the early trafficking of HIV-1 envelope protein through the secretory pathway. Overexpression of TBC1D20 hampered envelope processing and reduced its association with detergent-resistant membranes, entailing a reduction in infectivity of HIV-1 virion like particles (VLPs). CONCLUSIONS: These findings add TBC1D20 to the network of host factors regulating HIV replication cycle.


Assuntos
HIV-1/patogenicidade , Interações Hospedeiro-Patógeno , Replicação Viral , Produtos do Gene env do Vírus da Imunodeficiência Humana/metabolismo , Proteínas rab1 de Ligação ao GTP/metabolismo , Linhagem Celular , Humanos , Transporte Proteico
18.
J Virol ; 85(13): 6594-609, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21525358

RESUMO

YXXL motifs in cellular and viral proteins have a variety of functions. The matrix (M) protein of the respiratory pathogen human metapneumovirus (hMPV) contains two such conserved motifs--YSKL and YAGL. We mutated these sequences to analyze their contributions to hMPV infectivity. The mutant clones were capable of intracellular replication; however, the YAGL but not YSKL mutants were defective at spreading in infected cultures. We improved the reverse genetics system for hMPV and generated cell lines that stably expressed selectable, replicating full-length genomes for both the wild type and the mutant clones, allowing microscopic and biochemical analyses of these viruses. YAGL mutants produced normal cellular levels of M protein but failed to release virions, while ectopic coexpression of wild-type M generated particles that were restricted to a single cycle of infection. The YAGL motif did not act as a late (L) domain, however, since hMPV budding was independent of the cellular endosomal sorting complex required for transport (ESCRT) machinery and because replacement of the YAGL motif with classical L domains generated defective viruses. Instead, the YAGL mutants had defective M assemblies lacking a normal filamentous appearance and showed poor extractability from the cell compared to the wild-type protein. The mutant proteins were not grossly misfolded, however, as they interacted with cellular membranes and coassembled with wild-type M proteins. Thus, the YAGL motif is an important determinant of hMPV assembly. Furthermore, the selectable hMPV genomes described here should extend the use of reverse genetics systems in the analysis of spreading-defective viruses.


Assuntos
Motivos de Aminoácidos/genética , Metapneumovirus/patogenicidade , Proteínas da Matriz Viral/metabolismo , Vírion/metabolismo , Montagem de Vírus , Sequência de Aminoácidos , Animais , Linhagem Celular , Chlorocebus aethiops , Humanos , Metapneumovirus/genética , Metapneumovirus/metabolismo , Dados de Sequência Molecular , Mutação , Alinhamento de Sequência , Células Vero , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/genética
19.
PLoS Pathog ; 6(11): e1001183, 2010 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-21085616

RESUMO

The p12 protein is a cleavage product of the Gag precursor of the murine leukemia virus (MLV). Specific mutations in p12 have been described that affect early stages of infection, rendering the virus replication-defective. Such mutants showed normal generation of genomic DNA but no formation of circular forms, which are markers of nuclear entry by the viral DNA. This suggested that p12 may function in early stages of infection but the precise mechanism of p12 action is not known. To address the function and follow the intracellular localization of the wt p12 protein, we generated tagged p12 proteins in the context of a replication-competent virus, which allowed for the detection of p12 at early stages of infection by immunofluorescence. p12 was found to be distributed to discrete puncta, indicative of macromolecular complexes. These complexes were localized to the cytoplasm early after infection, and thereafter accumulated adjacent to mitotic chromosomes. This chromosomal accumulation was impaired for p12 proteins with a mutation that rendered the virus integration-defective. Immunofluorescence demonstrated that intracellular p12 complexes co-localized with capsid, a known constituent of the MLV pre-integration complex (PIC), and immunofluorescence combined with fluorescent in situ hybridization (FISH) revealed co-localization of the p12 proteins with the incoming reverse transcribed viral DNA. Interactions of p12 with the capsid and with the viral DNA were also demonstrated by co-immunoprecipitation. These results imply that p12 proteins are components of the MLV PIC. Furthermore, a large excess of wt PICs did not rescue the defect in integration of PICs derived from mutant p12 particles, demonstrating that p12 exerts its function as part of this complex. Altogether, these results imply that p12 proteins are constituent of the MLV PIC and function in directing the PIC from the cytoplasm towards integration.


Assuntos
Produtos do Gene gag/química , Produtos do Gene gag/metabolismo , Vírus da Leucemia Murina/fisiologia , Leucemia Experimental/metabolismo , Infecções por Retroviridae/metabolismo , Infecções Tumorais por Vírus/metabolismo , Montagem de Vírus , Animais , Western Blotting , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Neoplasias Ósseas/virologia , Células Cultivadas , Cromossomos de Mamíferos/genética , DNA Viral/genética , DNA Viral/metabolismo , Fibroblastos/metabolismo , Fibroblastos/virologia , Imunofluorescência , Produtos do Gene gag/genética , Humanos , Imunoprecipitação , Hibridização in Situ Fluorescente , Rim/citologia , Rim/metabolismo , Rim/virologia , Leucemia Experimental/patologia , Leucemia Experimental/virologia , Camundongos , Mitose/fisiologia , Mutação/genética , Células NIH 3T3 , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Osteossarcoma/virologia , Filogenia , RNA Mensageiro/genética , Infecções por Retroviridae/patologia , Infecções por Retroviridae/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Infecções Tumorais por Vírus/patologia , Infecções Tumorais por Vírus/virologia , Replicação Viral
20.
Pharmaceuticals (Basel) ; 15(3)2022 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-35337174

RESUMO

Controlling the infectivity of respiratory RNA viruses is critical, especially during the current SARS-CoV-2 pandemic. There is an unmet need for therapeutic agents that can reduce viral replication, preferably independent of the accumulation of viral mutations. Zinc ions have an apparent activity as modulators of intracellular viral RNA replication and thus, appear attractive in reducing viral RNA load and infectivity. However, the intracellular concentration of zinc is usually too low for achieving an optimal inhibitory effect. Various herbal polyphenols serve as excellent zinc ionophores with known antiviral properties. Here, we combined zinc picolinate with a collection of flavonoids, representing commonly used polyphenols. Copper was added to avoid ionic imbalance during treatment and to improve efficacy. Each component separately, as well as their combinations, did not interfere with the viability of cultured A549, H1299, or Vero cells in vitro as determined by MTT assay. The safe combinations were further evaluated to determine antiviral activity. Fluorescence-activated cell sorting and quantitative polymerase chain reaction were used to evaluate antiviral activity of the combinations. They revealed a remarkable (50-95%) decrease, in genome replication levels of a diverse group of respiratory RNA viruses, including the human coronavirus OC43 (HCoV-OC43; a betacoronavirus that causes the common cold), influenza A virus (IAV, strain A/Puerto Rico/8/34 H1N1), and human metapneumovirus (hMPV). Collectively, our results offer an orally bioavailable therapeutic approach that is non-toxic, naturally sourced, applicable to numerous RNA viruses, and potentially insensitive to new mutations and variants.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA