Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Biomolecules ; 11(4)2021 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-33921883

RESUMO

Parvovirus B19 (B19V) is a widespread human pathogen possessing a high tropism for erythroid precursor cells. However, the persistence or active replication of B19V in endothelial cells (EC) has been detected in diverse human pathologies. The VP1 unique region (VP1u) of the viral capsid has been reported to act as a major determinant of viral tropism for erythroid precursor cells. Nevertheless, the interaction of VP1u with EC has not been studied. We demonstrate that recombinant VP1u is efficiently internalized by rats' pulmonary trunk blood vessel-derived EC in vitro compared to the human umbilical vein EC line. The exposure to VP1u was not acutely cytotoxic to either human- or rat-derived ECs, but led to the upregulation of cellular stress signaling-related pathways. Our data suggest that high levels of circulating B19V during acute infection can cause endothelial damage, even without active replication or direct internalization into the cells.


Assuntos
Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Parvovirus B19 Humano/química , Proteínas Virais de Fusão/farmacologia , Animais , Sobrevivência Celular , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Artéria Pulmonar/citologia , Ratos , Ratos Wistar , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Proteínas Virais de Fusão/química
2.
Front Immunol ; 11: 1673, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32849580

RESUMO

Respiratory syncytial virus (RSV) remains the most common cause of lower respiratory tract infections in children worldwide. Development of a vaccine has been hindered by the risk of developing enhanced respiratory disease (ERD) upon natural exposure to the virus. Generation of higher quality neutralizing antibodies with stabilized pre-fusion F protein antigens has been proposed as a strategy to prevent ERD. We sought to test whether there was evidence of ERD in naïve BALB/c mice immunized with an unadjuvanted, stabilized pre-fusion F protein, and challenged with RSV line 19. We further sought to determine the extent to which formulation with a Th2-biased (alum) or a more Th1/Th2-balanced (Advax-SM) adjuvant influenced cellular responses and lung pathology. When exposed to RSV, mice immunized with pre-fusion F protein alone (PreF) exhibited increased airway eosinophilia and mucus accumulation. This was further exacerbated by formulation of PreF with Alum (aluminum hydroxide). Conversely, formulation of PreF with a Th1/Th2-balanced adjuvant, Advax-SM, not only suppressed RSV viral replication, but also inhibited airway eosinophilia and mucus accumulation. This was associated with lower numbers of lung innate lymphocyte cells (ILC2s) and CD4+ T cells producing IL-5+ or IL-13+ and increased IFNγ+ CD4+ and CD8+ T cells, in addition to RSV F-specific CD8+ T cells. These data suggest that in the absence of preimmunity, stabilized PreF antigens may still be associated with aberrant Th2 responses that induce lung pathology in response to RSV infection, and can be prevented by formulation with more Th1/Th2-balanced adjuvants that enhance CD4+ and CD8+ IFNγ+ T cell responses. This may support the use of stabilized PreF antigens with Th1/Th2-balanced adjuvants like, Advax-SM, as safer alternatives to alum in RSV vaccine candidates.


Assuntos
Adjuvantes Imunológicos/farmacologia , Hidróxido de Alumínio/farmacologia , Pulmão/efeitos dos fármacos , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/farmacologia , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Células Th2/efeitos dos fármacos , Proteínas Virais de Fusão/farmacologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Imunidade Humoral/efeitos dos fármacos , Imunização , Imunogenicidade da Vacina/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Camundongos Endogâmicos BALB C , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/patologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/imunologia , Vírus Sinciciais Respiratórios/patogenicidade , Equilíbrio Th1-Th2/efeitos dos fármacos , Células Th2/imunologia , Células Th2/metabolismo , Células Th2/virologia
3.
J Virol ; 94(13)2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32321812

RESUMO

Rabies virus (RABV) causes a severe and fatal neurological disease, but morbidity is vaccine preventable and treatable prior to the onset of clinical symptoms. However, immunoglobulin (IgG)-based rabies postexposure prophylaxis (PEP) is expensive, restricting access to life-saving treatment, especially for patients in low-income countries where the clinical need is greatest, and does not confer cross-protection against newly emerging phylogroup II lyssaviruses. Toward identifying a cost-effective replacement for the IgG component of rabies PEP, we developed and implemented a high-throughput screening protocol utilizing a single-cycle RABV reporter strain. A large-scale screen and subsequent direct and orthogonal counterscreens identified a first-in-class direct-acting RABV inhibitor, GRP-60367, with a specificity index (SI) of >100,000. Mechanistic characterization through time-of-addition studies, transient cell-to-cell fusion assays, and chimeric vesicular stomatitis virus (VSV) recombinants expressing the RABV glycoprotein (G) demonstrated that GRP-60367 inhibits entry of a subset of RABV strains. Resistance profiling of the chemotype revealed hot spots in conserved hydrophobic positions of the RABV G protein fusion loop that were confirmed in transient cell-to-cell fusion assays. Transfer of RABV G genes with signature resistance mutations into a recombinant VSV backbone resulted in the recovery of replication-competent virions with low susceptibility to the inhibitor. This work outlines a tangible strategy for mechanistic characterization and resistance profiling of RABV drug candidates and identified a novel, well-behaved molecular probe chemotype that specifically targets the RABV G protein and prevents G-mediated viral entry.IMPORTANCE Rabies PEP depends on anti-RABV IgG, which is expensive and in limited supply in geographical areas with the highest disease burden. Replacing the IgG component with a cost-effective and shelf-stable small-molecule antiviral could address this unmet clinical need by expanding access to life-saving medication. This study has established a robust protocol for high-throughput anti-RABV drug screens and identified a chemically well-behaved, first-in-class hit with nanomolar anti-RABV potency that blocks RABV G protein-mediated viral entry. Resistance mapping revealed a druggable site formed by the G protein fusion loops that has not previously emerged as a target for neutralizing antibodies. Discovery of this RABV entry inhibitor establishes a new molecular probe to advance further mechanistic and structural characterization of RABV G that may aid in the design of a next-generation clinical candidate against RABV.


Assuntos
Anticorpos Neutralizantes/uso terapêutico , Avaliação Pré-Clínica de Medicamentos/métodos , Vírus da Raiva/imunologia , Animais , Anticorpos Antivirais/imunologia , Antivirais/farmacologia , Linhagem Celular , Proteção Cruzada , Humanos , Biblioteca de Peptídeos , Raiva/prevenção & controle , Vacina Antirrábica/imunologia , Vírus da Raiva/metabolismo , Vírus da Raiva/patogenicidade , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/imunologia , Vesiculovirus/genética , Vesiculovirus/imunologia , Proteínas Virais de Fusão/farmacologia
4.
Vet Microbiol ; 235: 295-300, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31383316

RESUMO

Tembusu virus (TMUV), a member of the genus flavivirus, primarily causes egg-drop syndrome in ducks and is associated with low disease mortality but high morbidity. The commercially available live vaccines for treating TMUV currently include the main WF100, HB, and FX2010-180P strains, and efficient treatment and/or preventative measures are still urgently needed. Capsid-targeted viral inactivation (CTVI) is a conceptually powerful new antiviral strategy that is based on two proteins from the capsid protein of a virus and a crucial effector molecule. The effector molecule can destroy the viral DNA/RNA or interfere with the proper folding of key viral proteins, while the capsid protein mainly plays a role in viral integration and assembly; the fusion proteins are incorporated into virions during packaging. This study aimed to explore the potential use of this strategy in duck TMUV. Our results revealed that these fusion proteins can be expressed in susceptible BHK21 cells without cytotoxicity and possess excellent Ca2+-dependent nuclease activity, and their expression is also detectable in DF-1 cells. Compared to those in the negative controls (BHK21 and BHK21/pcDNA3.1(+) cells), the numbers of viral RNA copies in TMUV-infected BHK21/Cap-SNase and BHK21/Cap-Linker-SNase cells were reduced by 48 h, and the effect of Cap-Linker-SNase was superior to that of Cap-SNase. As anticipated, these results suggest that these fusion proteins contribute to viral resistance to treatment. Thus, CTVI might be applicable for TMUV inhibition as a novel antiviral therapeutic candidate during viral infection.


Assuntos
Proteínas do Capsídeo/farmacologia , Nuclease do Micrococo/farmacologia , Proteínas Virais de Fusão/farmacologia , Inativação de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Animais , Proteínas do Capsídeo/genética , Linhagem Celular , Patos , Flavivirus , Infecções por Flavivirus/tratamento farmacológico , Infecções por Flavivirus/virologia , Nuclease do Micrococo/genética
5.
J Am Chem Soc ; 141(32): 12648-12656, 2019 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-31268705

RESUMO

Human parainfluenza virus 3 (HPIV3) and respiratory syncytial virus (RSV) cause lower respiratory infection in infants and young children. There are no vaccines for these pathogens, and existing treatments have limited or questionable efficacy. Infection by HPIV3 or RSV requires fusion of the viral and cell membranes, a process mediated by a trimeric fusion glycoprotein (F) displayed on the viral envelope. Once triggered, the pre-fusion form of F undergoes a series of conformational changes that first extend the molecule to allow for insertion of the hydrophobic fusion peptide into the target cell membrane and then refold the trimeric assembly into an energetically stable post-fusion state, a process that drives the merger of the viral and host cell membranes. Peptides derived from defined regions of HPIV3 F inhibit infection by HPIV3 by interfering with the structural transitions of the trimeric F assembly. Here we describe lipopeptides derived from the C-terminal heptad repeat (HRC) domain of HPIV3 F that potently inhibit infection by both HPIV3 and RSV. The lead peptide inhibits RSV infection as effectively as does a peptide corresponding to the RSV HRC domain itself. We show that the inhibitors bind to the N-terminal heptad repeat (HRN) domains of both HPIV3 and RSV F with high affinity. Co-crystal structures of inhibitors bound to the HRN domains of HPIV3 or RSV F reveal remarkably different modes of binding in the N-terminal segment of the inhibitor.


Assuntos
Lipopeptídeos/farmacologia , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Inibidores de Proteínas Virais de Fusão/farmacologia , Proteínas Virais de Fusão/farmacologia , Sequência de Aminoácidos , Cristalografia por Raios X , Humanos , Lipopeptídeos/metabolismo , Testes de Sensibilidade Microbiana , Vírus da Parainfluenza 3 Humana/química , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Mucosa Respiratória/virologia , Vírus Sinciciais Respiratórios/química , Inibidores de Proteínas Virais de Fusão/metabolismo , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus/efeitos dos fármacos
6.
Fish Shellfish Immunol ; 85: 99-105, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29969707

RESUMO

The highly effective DNA vaccines against diseases caused by fish rhabdoviruses in farmed fish consist of a DNA plasmid vector encoding the viral glycoprotein under the control of a constitutive cytomegalovirus promoter (CMV). Among others, attempts to improve efficacy and safety of these DNA vaccines have focused on regulatory elements of plasmid vectors, which play a major role in controlling expression levels of vaccine antigens. Depending on the context, use of a fish-derived promoter with minimal activity in mammalian cells could be preferable. Another aspect related to the CMV promoter is that constitutive expression of the vaccine antigen may lead to rapid elimination of antigen expressing cells in the fish and thereby potentially reduce the long-term effects of the vaccine. In this study, we compared DNA vaccines with the interferon-inducible Mx promoter from rainbow trout and the CMV promoter, respectively. Plasmid constructs encoding the enhanced green fluorescent protein (EGFP) were used for the in vitro analysis, whereas DNA vaccines encoding the glycoprotein (G) of the viral haemorrhagic septicaemia virus (VHSV) were applied for the in vivo examination. The in vitro analysis showed that while the DNA vaccine with the CMV promoter constitutively drove the expression of EGFP in both fish and human cell lines, the DNA vaccine with the Mx promoter inducibly enhanced the expression of EGFP in the fish cell line. To address the impact on protection, a time-course model was followed as suggested by Kurath et al. (2006), where vaccinated fish were challenged with VHSV at 2, 8 and 78 weeks post-vaccination (wpv). The DNA vaccine with the CMV promoter protected at all times, while vaccination with the DNA vaccine containing the Mx promoter only protected the fish at 8 wpv. However, following induction with Poly (I:C) one week before the challenge, high protection was also evident at 2 wpv. In conclusion, the results revealed a more fish host dependent activity of the trout Mx promoter compared to the traditionally used cross species-active CMV promoter, but improvements will be needed for its application in DNA vaccines to ensure long term protection.


Assuntos
Doenças dos Peixes/prevenção & controle , Septicemia Hemorrágica Viral/prevenção & controle , Novirhabdovirus/imunologia , Oncorhynchus mykiss , Vacinas de DNA/farmacologia , Vacinas Virais/farmacologia , Animais , Linhagem Celular , Cyprinidae , Feminino , Doenças dos Peixes/imunologia , Doenças dos Peixes/virologia , Células HeLa , Septicemia Hemorrágica Viral/imunologia , Septicemia Hemorrágica Viral/virologia , Humanos , Interferons/imunologia , Perciformes , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Vacinas de DNA/administração & dosagem , Proteínas Virais de Fusão/administração & dosagem , Proteínas Virais de Fusão/farmacologia , Vacinas Virais/administração & dosagem
7.
Artigo em Inglês | MEDLINE | ID: mdl-29535974

RESUMO

The deep hydrophobic pocket of HIV-1 gp41 has been considered a drug target, but short-peptides targeting this site usually lack potent antiviral activity. By applying the M-T hook structure, we previously generated highly potent short-peptide fusion inhibitors that specifically targeted the pocket site, such as MT-SC22EK, HP23L, and LP-11. Here, the crystal structures of HP23L and LP-11 bound to the target mimic peptide N36 demonstrated the critical intrahelical and interhelical interactions, especially verifying that the hook-like conformation was finely adopted while the methionine residue was replaced by the oxidation-less prone residue leucine, and that addition of an extra glutamic acid significantly enhanced the binding and inhibitory activities. The structure of HP23L bound to N36 with two mutations (E49K and L57R) revealed the critical residues and motifs mediating drug resistance and provided new insights into the mechanism of action of inhibitors. Therefore, the present data help our understanding for the structure-activity relationship (SAR) of HIV-1 fusion inhibitors and facilitate the development of novel antiviral drugs.


Assuntos
Farmacorresistência Viral , Proteína gp41 do Envelope de HIV/química , Inibidores da Fusão de HIV/química , HIV-1 , Proteínas Virais de Fusão/química , Sequência de Aminoácidos , Substituição de Aminoácidos , Antivirais , Cristalografia por Raios X , Sistemas de Liberação de Medicamentos , Desenho de Fármacos , Ácido Glutâmico , Células HEK293 , Proteína gp41 do Envelope de HIV/genética , Proteína gp41 do Envelope de HIV/farmacologia , Inibidores da Fusão de HIV/farmacologia , HIV-1/química , HIV-1/genética , Humanos , Leucina , Modelos Moleculares , Mutação de Sentido Incorreto , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Peptídeos/química , Peptídeos/genética , Peptídeos/farmacologia , Conformação Proteica , Domínios Proteicos/genética , Domínios e Motivos de Interação entre Proteínas , Proteínas dos Retroviridae/genética , Proteínas dos Retroviridae/farmacologia , Relação Estrutura-Atividade , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/farmacologia
8.
EMBO Mol Med ; 10(2): 175-187, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29217660

RESUMO

Human respiratory syncytial virus (hRSV) and human metapneumovirus (hMPV), two members of the Pneumoviridae family, account for the majority of severe lower respiratory tract infections worldwide in very young children. They are also a frequent cause of morbidity and mortality in the elderly and immunocompromised adults. High levels of neutralizing antibodies, mostly directed against the viral fusion (F) glycoprotein, correlate with protection against either hRSV or hMPV However, no cross-neutralization is observed in polyclonal antibody responses raised after virus infection or immunization with purified F proteins. Based on crystal structures of hRSV F and hMPV F, we designed chimeric F proteins in which certain residues of well-characterized antigenic sites were swapped between the two antigens. The antigenic changes were monitored by ELISA with virus-specific monoclonal antibodies. Inoculation of mice with these chimeras induced polyclonal cross-neutralizing antibody responses, and mice were protected against challenge with the virus used for grafting of the heterologous antigenic site. These results provide a proof of principle for chimeric fusion proteins as single immunogens that can induce cross-neutralizing antibody and protective responses against more than one human pneumovirus.


Assuntos
Anticorpos Neutralizantes/imunologia , Metapneumovirus , Infecções por Paramyxoviridae , Proteínas Recombinantes de Fusão/imunologia , Infecções por Vírus Respiratório Sincicial , Vírus Sincicial Respiratório Humano , Proteínas Virais de Fusão/imunologia , Animais , Humanos , Imunização , Metapneumovirus/efeitos dos fármacos , Metapneumovirus/imunologia , Camundongos , Modelos Animais , Infecções por Paramyxoviridae/tratamento farmacológico , Infecções por Paramyxoviridae/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Vírus Sincicial Respiratório Humano/imunologia , Vacinas Sintéticas , Proteínas Virais de Fusão/farmacologia , Vacinas Virais
9.
Artigo em Inglês | MEDLINE | ID: mdl-28289031

RESUMO

Phage-derived lytic proteins are a promising alternative to conventional antimicrobials. One of their most interesting properties is that they do not readily select for resistant strains, which is likely due to the fact that their targets are essential for the viability of the bacterial cell. Moreover, genetic engineering allows the design of new "tailor-made" proteins that may exhibit improved antibacterial properties. One example of this is the chimeric protein CHAPSH3b, which consists of a catalytic domain from the virion-associated peptidoglycan hydrolase of phage vB_SauS-phiIPLA88 (HydH5) and the cell wall binding domain of lysostaphin. CHAPSH3b had previously shown the ability to kill Staphylococcus aureus cells. Here, we demonstrate that this lytic protein also has potential for the control of biofilm-embedded S. aureus cells. Additionally, subinhibitory doses of CHAPSH3b can decrease biofilm formation by some S. aureus strains. Transcriptional analysis revealed that exposure of S. aureus cells to this enzyme leads to the downregulation of several genes coding for bacterial autolysins. One of these proteins, namely, the major autolysin AtlA, is known to participate in staphylococcal biofilm development. Interestingly, an atl mutant strain did not display inhibition of biofilm development when grown at subinhibitory concentrations of CHAPSH3b, contrary to the observations made for the parental and complemented strains. Also, deletion of atl led to low-level resistance to CHAPSH3b and the endolysin LysH5. Overall, our results reveal new aspects that should be considered when designing new phage-derived lytic proteins aimed for antimicrobial applications.


Assuntos
Antibacterianos/farmacologia , N-Acetil-Muramil-L-Alanina Amidase/genética , Fagos de Staphylococcus/genética , Staphylococcus aureus/metabolismo , Staphylococcus aureus/virologia , Proteínas Virais de Fusão/farmacologia , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Parede Celular/metabolismo , Endopeptidases/metabolismo , Lisostafina/metabolismo , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/genética , Proteínas Virais de Fusão/genética
10.
Sci Rep ; 6: 28672, 2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27349522

RESUMO

During the co-evolution of viruses and their hosts, the viruses have evolved numerous strategies to counter and evade host antiviral immune responses in order to establish a successful infection, replicate and persist in the host. Recently, based on our model of immune signaling, the Signaling Chain HOmoOLigomerization (SCHOOL) model, we suggested specific molecular mechanisms used by different viruses such as severe acute respiratory syndrome coronavirus (SARS-CoV) to modulate the host immune response mediated by members of the family of multichain immune recognition receptors (MIRRs). This family includes T cell receptor (TCR) that is critically involved in immune diseases such as autoimmune arthritis. In the present study, we provide compelling experimental in vivo evidence in support of our hypothesis. Using the SCHOOL approach and the SARS-CoV fusion peptide sequence, we rationally designed a novel immunomodulatory peptide that targets TCR. We showed that this peptide ameliorates collagen-induced arthritis in DBA/1J mice and protects against bone and cartilage damage. Incorporation of the peptide into self-assembling lipopeptide nanoparticles that mimic native human high density lipoproteins significantly increases peptide dosage efficacy. Together, our data further confirm that viral immune evasion strategies that target MIRRs can be transferred to therapeutic strategies that require similar functionalities.


Assuntos
Artrite Experimental/tratamento farmacológico , Peptídeos/farmacologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/química , Proteínas Virais de Fusão/farmacologia , Animais , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Masculino , Camundongos , Peptídeos/química , Proteínas Virais de Fusão/química
11.
Chem Biol Drug Des ; 86(4): 487-95, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25581878

RESUMO

The emergence and dissemination of antibiotic-resistant bacterial pathogens have spurred the urgent need to develop novel antimicrobial agents with different mode of action. In this respect, we turned several fusogenic peptides (FPs) derived from the hemagglutinin glycoproteins (HAs) of IAV into potent antibacterials by replacing the negatively or neutrally charged residues of FPs with positively charged lysines. Their antibacterial activities were evaluated by testing the MICs against a panel of bacterial strains including S. aureus, S. mutans, P. aeruginosa, and E. coli. The results showed that peptides HA-FP-1, HA-FP-2-1, and HA-FP-3-1 were effective against both Gram-positive and Gram-negative bacteria with MICs ranging from 1.9 to 16.0 µm, while the toxicities toward mammalian cells were low. In addition, the mode of action and the secondary structure of these peptides were also discussed. These data not only provide several potent peptides displaying promising potential in development as broad antimicrobial agents, but also present a useful strategy in designing new antimicrobial agents.


Assuntos
Anti-Infecciosos , Bactérias/crescimento & desenvolvimento , Vírus da Influenza A/química , Peptídeos , Proteínas Virais de Fusão , Anti-Infecciosos/química , Anti-Infecciosos/farmacologia , Peptídeos/química , Peptídeos/farmacologia , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/farmacologia
12.
Biochim Biophys Acta ; 1848(2): 392-407, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25450346

RESUMO

The interaction between a peptide sequence from GB virus C E1 protein (E1P8) and its structural analogs (E1P8-12), (E1P8-13), and (E1P8-21) with anionic lipid membranes (POPG vesicles and POPG, DPPG or DPPC/DPPG (2:1) monolayers) and their association with HIV-1 fusion peptide (HIV-1 FP) inhibition at the membrane level were studied using biophysical methods. All peptides showed surface activity but leakage experiments in vesicles as well as insertion kinetics in monolayers and lipid/peptide miscibility indicated a low level of interaction: neither E1P8 nor its analogs induced the release of vesicular content and the exclusion pressure values (πe) were clearly lower than the biological membrane pressure (24-30 mN m(-1)) and the HIV-1 FP (35 mN m(-1)). Miscibility was elucidated in terms of the additivity rule and excess free energy of mixing (GE). E1P8, E1P8-12 and E1P8-21 (but not E1P8-13) induced expansion of the POPG monolayer. The mixing process is not thermodynamically favored as the positive GE values indicate. To determine how E1 peptides interfere in the action of HIV-1 FP at the membrane level, mixed monolayers of HIV-1 FP/E1 peptides (2:1) and POPG were obtained. E1P8 and its derivative E1P8-21 showed the greatest HIV-1 FP inhibition. The LC-LE phase lipid behavior was morphologically examined via fluorescence microscopy (FM) and atomic force microscopy (AFM). Images revealed that the E1 peptides modify HIV-1 FP-lipid interaction. This fact may be attributed to a peptide/peptide interaction as indicated by AFM results. Finally, hemolysis assay demonstrated that E1 peptides inhibit HIV-1 FP activity.


Assuntos
Vírus GB C/química , HIV-1/química , Bicamadas Lipídicas/química , Proteínas do Envelope Viral/química , Proteínas Virais de Fusão/química , Materiais Biomiméticos , Eritrócitos/citologia , Eritrócitos/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Humanos , Cinética , Membranas Artificiais , Fosfatidilgliceróis/química , Termodinâmica , Proteínas do Envelope Viral/farmacologia , Proteínas Virais de Fusão/antagonistas & inibidores , Proteínas Virais de Fusão/farmacologia
13.
Lab Chip ; 14(15): 2783-90, 2014 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-24898933

RESUMO

We present a microfluidic cell pairing device capable of sequential trapping and pairing of hundreds of cells using passive hydrodynamics and flow-induced deformation. We describe the design and operation principles of our device and show its applicability for cell fusion. Using our device, we achieved both homotypic and heterotypic cell pairing, demonstrating efficiencies up to 80%. The platform is compatible with fusion protocols based on biological, chemical and physical stimuli with fusion yields up to 95%. Our device further permits its disconnection from the fluidic hardware enabling its transportation for imaging and culture while maintaining cell registration on chip. Our design principles and cell trapping technique can readily be applied for different cell types and can be extended to trap and fuse multiple (>2) cell partners as demonstrated by our preliminary experiments.


Assuntos
Fibroblastos/citologia , Células Híbridas/citologia , Leucócitos/citologia , Técnicas Analíticas Microfluídicas/instrumentação , Animais , Fusão Celular , Linhagem Celular , Forma Celular , Tamanho Celular , Estimulação Elétrica , Desenho de Equipamento , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células Híbridas/efeitos dos fármacos , Células Híbridas/metabolismo , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Teste de Materiais , Camundongos , Microscopia de Fluorescência , Maleabilidade , Polietilenoglicóis/farmacologia , Impressão Tridimensional , Proteínas Recombinantes/metabolismo , Imagem com Lapso de Tempo , Proteínas Virais de Fusão/farmacologia
14.
J Clin Invest ; 124(5): 2113-24, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24743147

RESUMO

Respiratory syncytial virus (RSV) infection accounts for approximately 64 million cases of respiratory disease and 200,000 deaths worldwide each year, yet no broadly effective prophylactic or treatment regimen is available. RSV deploys paired, self-associating, heptad repeat domains of its fusion protein, RSV-F, to form a fusogenic 6-helix bundle that enables the virus to penetrate the host cell membrane. Here, we developed hydrocarbon double-stapled RSV fusion peptides that exhibit stabilized α-helical structure and striking proteolytic resistance. Pretreatment with double-stapled RSV peptides that specifically bound to the RSV fusion bundle inhibited infection by both laboratory and clinical RSV isolates in cells and murine infection models. Intranasal delivery of a lead double-stapled RSV peptide effectively prevented viral infection of the nares. A chitosan-based nanoparticle preparation markedly enhanced pulmonary delivery, further preventing progression of RSV infection to the lung. Thus, our results provide a strategy for inhibiting RSV infection by mucosal and endotracheal delivery of double-stapled RSV fusion peptides.


Assuntos
Doenças Nasais/prevenção & controle , Peptídeos , Pneumonia Viral/prevenção & controle , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sinciciais Respiratórios , Proteínas Virais de Fusão , Animais , Linhagem Celular , Quitosana/química , Quitosana/farmacologia , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/química , Doenças Nasais/metabolismo , Doenças Nasais/patologia , Peptídeos/química , Peptídeos/farmacologia , Pneumonia Viral/metabolismo , Pneumonia Viral/patologia , Infecções por Vírus Respiratório Sincicial/metabolismo , Infecções por Vírus Respiratório Sincicial/patologia , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/farmacologia
15.
Mar Drugs ; 12(3): 1512-29, 2014 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-24633252

RESUMO

To determine whether cholera toxin B subunit and active peptide from shark liver (CTB-APSL) fusion protein plays a role in treatment of type 2 diabetic mice, the CTB-APSL gene was cloned and expressed in silkworm (Bombyx mori) baculovirus expression vector system (BEVS), then the fusion protein was orally administrated at a dose of 100 mg/kg for five weeks in diabetic mice. The results demonstrated that the oral administration of CTB-APSL fusion protein can effectively reduce the levels of both fasting blood glucose (FBG) and glycosylated hemoglobin (GHb), promote insulin secretion and improve insulin resistance, significantly improve lipid metabolism, reduce triglycerides (TG), total cholesterol (TC) and low density lipoprotein (LDL) levels and increase high density lipoprotein (HDL) levels, as well as effectively improve the inflammatory response of type 2 diabetic mice through the reduction of the levels of inflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). Histopathology shows that the fusion protein can significantly repair damaged pancreatic tissue in type 2 diabetic mice, significantly improve hepatic steatosis and hepatic cell cloudy swelling, reduce the content of lipid droplets in type 2 diabetic mice, effectively inhibit renal interstitial inflammatory cells invasion and improve renal tubular epithelial cell nucleus pyknosis, thus providing an experimental basis for the development of a new type of oral therapy for type 2 diabetes.


Assuntos
Toxina da Cólera/farmacologia , Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/farmacologia , Proteínas Virais de Fusão/farmacologia , Animais , Baculoviridae/genética , Baculoviridae/metabolismo , Glicemia/metabolismo , Peso Corporal , Bombyx/virologia , Toxina da Cólera/química , DNA Viral/genética , Diabetes Mellitus Experimental/tratamento farmacológico , Escherichia coli/genética , Escherichia coli/metabolismo , Gangliosídeo G(M1)/metabolismo , Vetores Genéticos , Hemoglobinas Glicadas/análise , Hipolipemiantes/farmacologia , Insulina/metabolismo , Resistência à Insulina , Rim/patologia , Lipídeos/sangue , Masculino , Camundongos , Camundongos Endogâmicos ICR , Tubarões , Baço/patologia , Proteínas Virais de Fusão/química
16.
J Virol Methods ; 199: 11-6, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24412629

RESUMO

Newcastle disease virus (NDV) of paramyxovirus and Marek's disease virus (MDV) of herpesvirus, two of the most serious threats to the poultry industry, can give rise to complex co-infections that hinder diagnosis and prevention. In the current study, two different peptides, derived from the MDV gH (gHH2L) and gB (gBH3), respectively, exhibit antiviral activity against NDV in vitro. The potent inhibitory effect of heptad repeat 2 from fusion glycoprotein of the NDV on MDV infection also has been demonstrated. Plaque formation and embryo infectivity assays confirmed these antiviral results. Furthermore, each tandem peptide consisting of two motifs from different viruses exhibits more potent antiviral activity than the constituent peptides. The current work provides a new strategy for developing novel peptides and vaccines against virus infection and co-infections.


Assuntos
Antivirais/farmacologia , Glicoproteínas/farmacologia , Mardivirus/efeitos dos fármacos , Vírus da Doença de Newcastle/efeitos dos fármacos , Peptídeos/farmacologia , Proteínas Virais de Fusão/farmacologia , Animais , Antivirais/uso terapêutico , Linhagem Celular , Embrião de Galinha , Glicoproteínas/genética , Glicoproteínas/uso terapêutico , Mardivirus/genética , Doença de Marek/prevenção & controle , Testes de Sensibilidade Microbiana , Doença de Newcastle/prevenção & controle , Vírus da Doença de Newcastle/genética , Peptídeos/genética , Peptídeos/uso terapêutico , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/uso terapêutico , Ensaio de Placa Viral
17.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 42(2): 152-6, 2011 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-21500543

RESUMO

OBJECTIVE: To observe the killing effect of recombinant type I herpes simplex virus (HSV-I) with Gibbon ape leukemia virus membrane fusion glycoprotein (GALV.fus) gene on lung adenocarcinoma in vitro and in vivo. METHODS: Recombinant HSV-I plasmids (HSV-UL38P-GALV.fus, HSV-CMVP-GALV.fus, HSV-CMVP-EGFP) was introduced into green monkey kidney cells (Vero) by liposome to amplify the virus, which were propagated in Vero cells and purified by cesium chloride density purification, titrated by TCID50 method. The three recombinant viruses were named as Synco-2, Synco-1 and Baco-1 respectively, and were transfected into lung adenocarcinoma cell line A549 cell and human lung adenocarcinoma xenografts which were established in nude mice subcutaneously to observe the expression and transfection of recombinant plasmids; mice model was divided to A (Control) group, B (Baco-1) group, C (Synco-1) group, D (Synco-2) group and E (Synco-2) group. The antitumor and cytotoxic effects of the virus in vitro or in vivo were investigated simultaneously. RESULTS: Recombinated HSV-I virus were packed successfully, the titre of Baco-1, Synco-1 and Synco-2 were 3 x 10(10) pfu/mL, 1X 10(11) pfu/mL and 4 x 10(10) pfu/mL respectively. The virus produced clear antitumor effects in vitro, the oncolytic activity of Synco-2 and Synco-1 was superior to that of Baco-1 (P < 0.01). The striking antitumor effect was seen when the virus was given subcutaneously in established xenografts in the animals. Tumor volume in Group C and D decreased significantly compared those in Group A and B (P < 0.01). The same result was observed in tumour weight (P < 0.01), and we also find that there was statistical significance between Group C and D in tumour quality at last two weeks (P < 0.01). CONCLUSIONS: The three recombinant HSV-I were packaged, amplificated and purified successfully. Recombinant GALV. fus gene system controlled by special promoter and mediated by available carrier has potent activity against lung cancer both in vitro or in vivo, and maybe a new promising candidate for investigative gene therapy of this malignancy.


Assuntos
Terapia Genética/métodos , Herpesvirus Humano 1/metabolismo , Vírus da Leucemia do Macaco Gibão/genética , Neoplasias Pulmonares/terapia , Glicoproteínas de Membrana/genética , Proteínas Virais de Fusão/farmacologia , Adenocarcinoma/patologia , Adenocarcinoma/terapia , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Herpesvirus Humano 1/genética , Humanos , Neoplasias Pulmonares/patologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Nus , Transplante de Neoplasias , Transfecção , Células Vero , Proteínas Virais de Fusão/genética
18.
Cell Immunol ; 263(2): 154-60, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20406704

RESUMO

A fusion protein of single chain antibody (scFv) specific for transferrin receptor (TfR, CD71) and viral peptide/HLA-A2 complex was prepared in this study to redirect cytotoxic T cells (CTLs) of viral specificity to tumor cells by attaching the ligand of T cell receptor (TCR) to tumor cells via binding of TfR scFv to TfR. The results demonstrate that the fusion protein can attach the active virus-peptide/HLA-A2 complex to HLA class I-negative, TfR-expressing K562 cells through binding of TfR scFv to TfR, and mediate cytotoxicity of viral peptide-specific CTLs against K562 cells in vitro. In addition, the fusion protein can induce inhibition of solid tumor formation and improve survival time in tumor xenograft nude mouse with the injection of the sorted viral peptide-specific CTLs generated by co-culture of peripheral blood lymphocytes from HLA-A2 positive donors with inactivated T2 cells pulsed with the viral peptide.


Assuntos
Antígenos de Neoplasias , Sobrevivência Celular/efeitos dos fármacos , Fragmentos de Peptídeos/imunologia , Receptores da Transferrina/imunologia , Linfócitos T Citotóxicos/imunologia , Proteínas Virais de Fusão/farmacologia , Animais , Antígenos de Neoplasias/imunologia , Linhagem Celular Tumoral , Feminino , Antígeno HLA-A2 , Humanos , Células K562 , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ligação Proteica , Receptores da Transferrina/genética , Linfócitos T Citotóxicos/citologia , Proteínas Virais de Fusão/imunologia
19.
J Infect Dis ; 201(8): 1201-7, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20205592

RESUMO

Human respiratory syncytial virus (RSV) is a ubiquitous respiratory virus that causes serious lower respiratory tract disease in infants and young children worldwide. Studies have shown that RSV infection modulates chemokine expression patterns, suggesting that particular cytokine expression profiles may be indicators of disease severity. In this study, we show that RSV F or G protein treatment of fully differentiated primary normal human bronchial epithelial cells induces apical and basolateral secretion of interleukin 8 (IL-8), interferon-inducible protein 10 (IP-10), monocyte chemotactic protein 1 (MCP-1), and RANTES (regulated on activation, normal T cell expressed and secreted). Purified RSV G (attachment) protein was shown to stimulate the secretion of interleukin 1alpha and RANTES, whereas purified F (fusion) protein elicited the production of IL-8, IP-10, and RANTES. Studies of ultraviolet-inactivated RSV showed that treatment of normal human bronchial epithelial cells induces apical IL-8, IP-10, and MCP-1 secretion independent of infection, suggesting that RSV proteins alone modify the chemokine response pattern, which may affect the early immune response before infection.


Assuntos
Brônquios/virologia , Quimiocinas CC/biossíntese , Quimiocinas CXC/biossíntese , Interleucina-1alfa/biossíntese , Proteínas Virais de Fusão/farmacologia , Adolescente , Brônquios/imunologia , Células Cultivadas , Quimiocina CXCL10/biossíntese , Epitélio/imunologia , Epitélio/virologia , Regulação Viral da Expressão Gênica/fisiologia , Humanos , Interleucina-8/biossíntese , Masculino , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia
20.
Expert Rev Anti Infect Ther ; 7(9): 1125-9, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19883332

RESUMO

Respiratory syncytial virus (RSV) is a pathogen whose existence has been known for decades, causing mild-to-severe upper and lower respiratory tract infections that bear the risk of subsequent asthma and can even lead to a fatal outcome. RSV infects all groups of patients and is a major cause of hospitalization in children and in the elderly. This review briefly summarizes the current status of RSV drug development and clinical trials for drugs available for the treatment of RSV infections.


Assuntos
Antivirais/uso terapêutico , Descoberta de Drogas , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Antivirais/farmacologia , Benzimidazóis/uso terapêutico , Ensaios Clínicos como Assunto , Glicosaminoglicanos/antagonistas & inibidores , Glicosaminoglicanos/farmacologia , Humanos , Receptores de Lipopolissacarídeos/metabolismo , RNA Interferente Pequeno/uso terapêutico , Proteínas Virais de Fusão/antagonistas & inibidores , Proteínas Virais de Fusão/farmacologia , Ligação Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...