Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Negl Trop Dis ; 16(5): e0010315, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35576193

RESUMO

Establishment of novel mosquito control technologies such as the use of genetically engineered insects typically involves phased testing to generate robust data-sets that support its safe and effective use as a vector control tool. In this study, we demonstrate the ability of the transgenic self-limiting OX513A Aedes aegypti strain to suppress a wild type Ae. aegypti population in an outdoor containment facility in India. OX513A is a genetically engineered Ae. aegypti strain with a repressible dominant self-limiting gene. When male adult OX513A mate with wild female adults, a single copy of the self-limiting gene is inherited by all the progeny, leading to death of >95% of progeny during larval/pupal development. A wild-type population of Ae. aegypti was established and stabilized during a 14 week period in five paired field cage units, each consisting of control and treatment cages, followed by weekly releases of OX513A male adults to suppress the target population. The successive introductions of OX513A male adults led to a consistent decline in wild type numbers eventually resulting in the elimination of Ae. aegypti from all treated cages within 10 to 15 weeks of release. This study demonstrates that Ae. aegypti elimination may be a realistic and achievable target in relatively isolated environments.


Assuntos
Aedes , Febre Amarela , Aedes/genética , Animais , Feminino , Masculino , Controle de Mosquitos , Mosquitos Vetores/genética , Controle Biológico de Vetores/métodos
2.
Proc Natl Acad Sci U S A ; 110(50): 20063-8, 2013 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-24277846

RESUMO

During infection, viruses undergo conformational changes that lead to delivery of their genome into host cytosol. In human rhinovirus A2, this conversion is triggered by exposure to acid pH in the endosome. The first subviral intermediate, the A-particle, is expanded and has lost the internal viral protein 4 (VP4), but retains its RNA genome. The nucleic acid is subsequently released, presumably through one of the large pores that open at the icosahedral twofold axes, and is transferred along a conduit in the endosomal membrane; the remaining empty capsids, termed B-particles, are shuttled to lysosomes for degradation. Previous structural analyses revealed important differences between the native protein shell and the empty capsid. Nonetheless, little is known of A-particle architecture or conformation of the RNA core. Using 3D cryo-electron microscopy and X-ray crystallography, we found notable changes in RNA-protein contacts during conversion of native virus into the A-particle uncoating intermediate. In the native virion, we confirmed interaction of nucleotide(s) with Trp(38) of VP2 and identified additional contacts with the VP1 N terminus. Study of A-particle structure showed that the VP2 contact is maintained, that VP1 interactions are lost after exit of the VP1 N-terminal extension, and that the RNA also interacts with residues of the VP3 N terminus at the fivefold axis. These associations lead to formation of a well-ordered RNA layer beneath the protein shell, suggesting that these interactions guide ordered RNA egress.


Assuntos
Modelos Moleculares , Conformação de Ácido Nucleico , RNA Viral/metabolismo , Rhinovirus/fisiologia , Vírion/química , Desenvelopamento do Vírus/fisiologia , Microscopia Crioeletrônica , Cristalografia por Raios X , Humanos , Processamento de Imagem Assistida por Computador , RNA Viral/química , Rhinovirus/genética
3.
Electrophoresis ; 33(12): 1833-41, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22740471

RESUMO

During infection, enteroviruses, such as human rhinoviruses (HRVs), convert from the native, infective form with a sedimentation coefficient of 150S to empty subviral particles sedimenting at 80S (B particles). B particles lack viral capsid protein 4 (VP4) and the single-stranded RNA genome. On the way to this end stage, a metastable intermediate particle is observed in the cell early after infection. This subviral A particle still contains the RNA but lacks VP4 and sediments at 135S. Native (150S) HRV serotype 2 (HRV2) as well as its empty (80S) capsid have been well characterized by capillary electrophoresis. In the present paper, we demonstrate separation of at least two forms of subviral A particles on the midway between native virions and empty 80S capsids by CE. For one of these intermediates, we established a reproducible way for its preparation and characterized this particle in terms of its electrophoretic mobility and its appearance in transmission electron microscopy (TEM). Furthermore, the conversion of this intermediate to 80S particles was investigated. Gas-phase electrophoretic mobility molecular analysis (GEMMA) yielded additional insights into sample composition. More data on particle characterization including its protein composition and RNA content (for unambiguous identification of the detected intermediate as subviral A particle) will be presented in the second part of the publication.


Assuntos
Eletroforese Capilar/métodos , Microscopia Eletrônica/métodos , Rhinovirus/química , Espectrometria de Massas por Ionização por Electrospray/métodos , Vírion/química , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Tamanho da Partícula , Temperatura , Tireoglobulina/química
4.
PLoS Pathog ; 8(1): e1002473, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22241997

RESUMO

Upon attachment to their respective receptor, human rhinoviruses (HRVs) are internalized into the host cell via different pathways but undergo similar structural changes. This ultimately results in the delivery of the viral RNA into the cytoplasm for replication. To improve our understanding of the conformational modifications associated with the release of the viral genome, we have determined the X-ray structure at 3.0 Å resolution of the end-stage of HRV2 uncoating, the empty capsid. The structure shows important conformational changes in the capsid protomer. In particular, a hinge movement around the hydrophobic pocket of VP1 allows a coordinated shift of VP2 and VP3. This overall displacement forces a reorganization of the inter-protomer interfaces, resulting in a particle expansion and in the opening of new channels in the capsid core. These new breaches in the capsid, opening one at the base of the canyon and the second at the particle two-fold axes, might act as gates for the externalization of the VP1 N-terminus and the extrusion of the viral RNA, respectively. The structural comparison between native and empty HRV2 particles unveils a number of pH-sensitive amino acid residues, conserved in rhinoviruses, which participate in the structural rearrangements involved in the uncoating process.


Assuntos
Proteínas do Capsídeo/química , Capsídeo/química , RNA Viral/química , Rhinovirus/química , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Cristalografia por Raios X , Humanos , Interações Hidrofóbicas e Hidrofílicas , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , RNA Viral/metabolismo , Rhinovirus/metabolismo , Internalização do Vírus
7.
J Virol ; 85(16): 8368-75, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21680510

RESUMO

After uptake into target cells, many nonenveloped viruses undergo conformational changes in the low-pH environment of the endocytic compartment. This results in exposure of amphipathic viral peptides and/or hydrophobic protein domains that are inserted into and either disrupt or perforate the vesicular membranes. The viral nucleic acids thereby gain access to the cytosol and initiate replication. We here demonstrate the in vitro transfer of the single-stranded positive-sense RNA genome of human rhinovirus 2 into liposomes decorated with recombinant very-low-density lipoprotein receptor fragments. Membrane-attached virions were exposed to pH 5.4, mimicking the in vivo pH environment of late endosomes. This triggered the release of the RNA whose arrival in the liposomal lumen was detected via in situ cDNA synthesis by encapsulated reverse transcriptase. Subsequently, cDNA was PCR amplified. At a low ratio between virions and lipids, RNA transfer was positively correlated with virus concentration. However, membranes became leaky at higher virus concentrations, which resulted in decreased cDNA synthesis. In accordance with earlier in vivo data, the RNA passes through the lipid membrane without causing gross damage to vesicles at physiologically relevant virus concentrations.


Assuntos
Membrana Celular/metabolismo , Lipossomos , Infecções por Vírus de RNA/virologia , Rhinovirus/fisiologia , Internalização do Vírus , Desenvelopamento do Vírus , Membrana Celular/virologia , Genoma Viral , Concentração de Íons de Hidrogênio , Lipídeos de Membrana/metabolismo , Microscopia Eletrônica de Transmissão , Nanoestruturas , RNA Viral/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Virology ; 412(1): 55-67, 2011 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-21262518

RESUMO

The major group human rhinovirus type 8 can enter cells via heparan sulphate. When internalized into ICAM-1 negative rhabdomyosarcoma (RD) cells, HRV8 accumulated in the cells but caused CPE only after 3 days when used at high MOI. Adaptation by three blind passages alternating between RD and HeLa cells resulted in variant HRV8v with decreased stability at acidic pH allowing for productive infection in the absence of ICAM-1. HRV8v produced CPE at 10 times lower MOI within 1 day. Confocal fluorescence microscopy colocalization and the use of pharmacological and dominant negative inhibitors revealed that viral uptake is clathrin, caveolin, and flotillin independent. However, it is blocked by dynasore, amiloride, and EIPA. Furthermore, HRV8v induced FITC-dextran uptake and colocalized with this fluid phase marker. Except for the complete inhibition by dynasore, the entry pathway of HRV8v via HS is similar to that of HRV14 in RD cells that overexpress ICAM-1.


Assuntos
Dinamina II/metabolismo , Interações Hospedeiro-Patógeno , Molécula 1 de Adesão Intercelular/metabolismo , Rhinovirus/fisiologia , Internalização do Vírus , Caveolinas/genética , Caveolinas/metabolismo , Linhagem Celular , Clatrina/genética , Clatrina/metabolismo , Efeito Citopatogênico Viral , Dinamina II/genética , Humanos , Molécula 1 de Adesão Intercelular/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Rhinovirus/genética , Inoculações Seriadas
9.
J Virol Methods ; 171(1): 206-11, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21056058

RESUMO

The genomic RNA of picornaviruses is attached to a small protein (VPg) via a covalent bond between a tyrosine and a 5'-terminal uridine phosphate. The same structure is present in potyvirus and calicivirus families. VPgs play a key role in initiation of viral replication by acting as primers for RNA synthesis. The model compound [N(Ac),CO(NHMe)]Tyr-(5'P→O)Up-O-(CH(2))(6)NH(2) (mCLU), mimicking this 'covalent linkage unit' (CLU) and containing Tyr-pUp was synthesized in solution following the phosphoramidite scheme and used to raise antibodies for studying picornavirus infection. The antibodies recognized CLU-containing mengovirus RNA and showed minimal cross-reactivity with RNAs lacking CLU. Immunofluorescence staining of cells infected with a human rhinovirus demonstrated co-localization of the signals from anti-mCLU and from anti-VPg antibodies. Efficient synthesis of mCLU and anti-mCLU antibodies might be of great utility for investigating viral replication and identifying yet unknown viral and cellular CLU-containing RNA-protein complexes.


Assuntos
Anticorpos Antivirais , Oligorribonucleotídeos/síntese química , Oligorribonucleotídeos/imunologia , Picornaviridae/crescimento & desenvolvimento , RNA Viral/análise , Virologia/métodos , Animais , Anticorpos Antivirais/isolamento & purificação , Células HeLa , Humanos , Microscopia de Fluorescência/métodos , Picornaviridae/química , Coelhos
10.
Anal Chem ; 82(19): 8146-52, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20806784

RESUMO

Permeabilization of model lipid membranes by virus-derived peptides, viral proteins, and entire virions of human rhinovirus was assessed by quantifying the release of a fluorescent dye from liposomes via a novel chip electrophoretic assay. Liposomal leakage readily occurred upon incubation with the pH-sensitive synthetic fusogenic peptide GALA and, less efficiently, with a 24mer peptide (P1-N) derived from the N-terminus of the capsid protein VP1 of human rhinovirus 2 (HRV2) at acidic pH. Negative stain transmission electron microscopy showed that liposomes incubated with the rhinovirus-derived peptide remained largely intact. At similar concentrations, the GALA peptide caused gross morphological changes of the liposomes. On a molar basis, the leakage-inducing efficiency of the P1 peptide was by about 2 orders of magnitude inferior to that of recombinant VP1 (from HRV89) and entire HRV2. This underscores the role in membrane destabilization of VP1 domains remote from the N-terminus and the arrangement of the peptide in the context of the icosahedral virion. Our method is rapid, requires tiny amounts of sample, and allows for the parallel determination of released and retained liposomal cargo.


Assuntos
Eletroforese/métodos , Lipossomos/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Peptídeos/metabolismo , Proteínas Virais/metabolismo , Vírion/metabolismo , Sequência de Aminoácidos , Corantes Fluorescentes , Humanos , Microscopia Eletrônica de Transmissão , Rhinovirus/metabolismo
11.
J Virol ; 84(8): 3984-92, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20130060

RESUMO

Intercellular adhesion molecule 1 (ICAM-1) mediates binding and entry of major group human rhinoviruses (HRVs). Whereas the entry pathway of minor group HRVs has been studied in detail and is comparatively well understood, the pathway taken by major group HRVs is largely unknown. Use of immunofluorescence microscopy, colocalization with specific endocytic markers, dominant negative mutants, and pharmacological inhibitors allowed us to demonstrate that the major group virus HRV14 enters rhabdomyosarcoma cells transfected to express human ICAM-1 in a clathrin-, caveolin-, and flotillin-independent manner. Electron microscopy revealed that many virions accumulated in long tubular structures, easily distinguishable from clathrin-coated pits and caveolae. Virus entry was strongly sensitive to the Na(+)/H(+) ion exchange inhibitor amiloride and moderately sensitive to cytochalasin D. Thus, cellular uptake of HRV14 occurs via a pathway exhibiting some, but not all, characteristics of macropinocytosis and is similar to that recently described for adenovirus 3 entry via alpha(v) integrin/CD46 in HeLa cells.


Assuntos
Molécula 1 de Adesão Intercelular/biossíntese , Células Musculares/virologia , Rhinovirus/fisiologia , Internalização do Vírus , Amilorida/farmacologia , Caveolinas/metabolismo , Linhagem Celular Tumoral , Clatrina/metabolismo , Citocalasina D/farmacologia , Humanos , Proteínas de Membrana/metabolismo , Microscopia Confocal , Microscopia Eletrônica , Microscopia de Fluorescência , Bloqueadores dos Canais de Sódio/farmacologia
12.
Electrophoresis ; 30(24): 4292-9, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20013915

RESUMO

In first attempts at elucidating the transfer of the RNA genome of a human Rhinovirus through lipid membranes in vitro we made use of liposomes decorated with recombinant receptors. This model system was characterized previously by CE but suffered from the requirement for inclusion of polyethylene glycol-modified lipids for reliable separations [Weiss, V. U., Bilek, G., Pickl-Herk, A., Blaas, D., Kenndler, E., Electrophoresis 2009, 30, 2123-2128.]. We here report the analysis of liposomes with a lipid composition much more similar to that of biological lipid bilayers. We found that vesicles containing and lacking this non-physiologic lipid differ significantly in their electrophoretic mobility (by factor 2) although the concentration of charge-bearing lipids in their bilayers is the same. We demonstrate that binding of a human Rhinovirus to the latter liposomes decorated with a cognate receptor can be analysed via electrophoresis on microchips; we support our results with transmission electron microscopy.


Assuntos
Eletroforese Capilar/métodos , Dispositivos Lab-On-A-Chip , Lipídeos/análise , Lipossomos , Modelos Biológicos , Viroses/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Microscopia Eletrônica de Transmissão
13.
J Virol ; 83(21): 10922-30, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19706701

RESUMO

Minor group human rhinoviruses (HRVs) bind three members of the low-density lipoprotein receptor (LDLR) family: LDLR proper, very-LDLR (VLDLR) and LDLR-related protein (LRP). Whereas ICAM-1, the receptor of major group HRVs actively contributes to viral uncoating, LDLRs are rather considered passive vehicles for cargo delivery to the low-pH environment of endosomes. Since the Tyr-Trp-Thr-Asp beta-propeller domain of LDLR has been shown to be involved in the dissociation of bound LDL via intramolecular competition at low pH, we studied whether it also plays a role in HRV infection. Human cell lines deficient in LDLR family proteins are not available. Therefore, we used CHO-ldla7 cells that lack endogenous LDLR. These were stably transfected to express either wild-type (wt) human LDLR or a mutant with a deletion of the beta-propeller. When HRV2 was attached to the propeller-negative LDLR, a lower pH was required for conversion to subviral particles than when attached to wt LDLR. This indicates that high-avidity receptor binding maintains the virus in its native conformation. HRV2 internalization directed the mutant LDLR but not wt LDLR to lysosomes, resulting in reduced plasma membrane expression of propeller-negative LDLR. Infection assays using a CHO-adapted HRV2 variant showed a delay in intracellular viral conversion and de novo viral synthesis in cells expressing the truncated LDLR. Our data indicate that the beta-propeller attenuates the virus-stabilizing effect of LDLR binding and thereby facilitates RNA release from endosomes, resulting in the enhancement of infection. This is a nice example of a virus exploiting high-avidity multimodule receptor binding with an intrinsic release mechanism.


Assuntos
Infecções por Picornaviridae/metabolismo , Estrutura Secundária de Proteína , Receptores de LDL/química , Receptores de LDL/metabolismo , Rhinovirus/fisiologia , Internalização do Vírus , Animais , Células CHO , Cricetinae , Cricetulus , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Lipoproteínas LDL/metabolismo , Lisossomos/metabolismo , Mutação , RNA Viral/genética , RNA Viral/metabolismo , Receptores de LDL/genética , Ligação Viral
14.
FEBS Lett ; 583(15): 2547-51, 2009 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-19615999

RESUMO

Major group HRVs bind intercellular adhesion molecule 1 and minor group HRVs bind members of the low-density lipoprotein receptor (LDLR) family for cell entry. Whereas the former share common sequence motives in their viral capsid proteins (VPs), in the latter only a lysine residue within the binding epitope in VP1 is conserved; this lysine is also present in "K-type" major group HRVs that fail to use LDLR for infection. By using the available sequences three-dimensional models of VP1 of all HRVs were built and binding energies, with respect to module 3 of the very-low-density lipoprotein receptor, were calculated. Based on the predicted affinities K-type HRVs and minor group HRVs were correctly classified.


Assuntos
Biologia Computacional , Rhinovirus/química , Rhinovirus/genética , Ligação Viral , Sequência de Aminoácidos , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Receptores de LDL/genética , Receptores de LDL/metabolismo , Rhinovirus/classificação , Rhinovirus/metabolismo , Software
15.
Electrophoresis ; 30(12): 2123-8, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19582713

RESUMO

Electrophoresis on a chip increasingly replaces electrophoresis in the capillary format because of its speed and containment of the sample within a disposable cartridge. In this paper we demonstrate its utility in the analysis of the interaction between a virus and a liposome-anchored receptor, mimicking viral attachment to host cells. This became possible because detergents, obligatory constituents of the BGE for capillary electrophoretic separation of the virus, were not necessary in the chip format. Separations were carried out in sodium borate buffer, pH 8.3. Liposomes and virus were both labeled for laser-induced fluorescence detection at lambda(ex)/lambda(em) 630/680 nm. Free virus and virus-receptor complexes were resolved from virus attached to receptor-decorated liposomes in the absence of additives or sieving matrices within about 30 s on commercially available microfluidic chips.


Assuntos
Eletroforese em Microchip/métodos , Lipossomos/metabolismo , Rhinovirus/fisiologia , Ligação Viral , Fluorescência , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Lipossomos/química , Receptores de LDL/metabolismo , Rhinovirus/isolamento & purificação , Rhinovirus/metabolismo
16.
Biochimie ; 91(7): 903-15, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19401214

RESUMO

Circulating 'free' non-enveloped Hepatitis C virus (HCV) core protein has been demonstrated in HCV-infected patients, and HCV subgenomes with deletions of the envelope proteins have been previously identified. Initial studies from our laboratory, previously published, indicated that expression of HCV core in insect cells can direct the formation of capsid-like particles lacking the envelope glycoproteins. These protein nanospheres, morphologically similar to natural capsids, were shown to be taken up by human hepatic cells and to produce cell-signalling events. To follow the intracellular fate of these particles we fused the core protein to eGFP. We demonstrate that the chimeric proteins core(173)-eGFP, eGFP-core(191) and eGFP-core(173) can be efficiently expressed, self-assembled, and form fluorescent non-enveloped capsid-like particles. By using confocal microscopy and FACS analysis, we provide evidence that the fluorescent nanospheres can not only enter human hepatic cells - the main target of HCV - but also human immune cells such as T and B lymphocytes, as well as human myeloid leukaemia cells differentiated along the monocyte/macrophage-like pathway. The fluorescent particles might thus be used to trace the intracellular trafficking of naked HCV capsids as showed by live microscopy and to further understand their biological significance.


Assuntos
Capsídeo/fisiologia , Proteínas de Fluorescência Verde/análise , Hepacivirus/fisiologia , Hepatócitos/virologia , Proteínas do Core Viral/análise , Internalização do Vírus , Linfócitos B/virologia , Capsídeo/ultraestrutura , Linhagem Celular Tumoral , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hepacivirus/ultraestrutura , Humanos , Monócitos/virologia , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Linfócitos T/virologia , Proteínas do Core Viral/genética , Proteínas do Core Viral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA