Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-38183648

RESUMO

BACKGROUND: Upper respiratory tract is the primary target of SARS-CoV-2. Therefore, nasal immune responses act as the first line of defense against SARS-CoV-2 infection. OBJECTIVE: We aim to investigate the immune responses of human nasal epithelial cells (HNEpCs) upon stimulation with a COVID-19 vaccine candidate. This candidate named RBD-NPs is composed of SARS-CoV-2 receptor-binding domain (RBD) encapsulated within the N,N,N-trimethyl chitosan nanoparticles (TMC-NPs). METHODS: HNEpCs were stimulated with RBD-NPs, empty NPs, or soluble RBD at various concentrations. After 24 and 48 h of treatment, cells viability and delivery of the immunogens were assessed using XTT assay and flow cytometry. Levels of cytokines and chemokines in the supernatant were quantified with Bio-plex Human Cytokine Assay. Communication between RBD-NPs-stimulated HNEpCs and monocyte-derived dendritic cells (MoDCs) was assessed through differentiation of MoDCs into mature phenotype. RESULTS: RBD-NPs as high as 100 µg exerted no toxicity to HNEpCs and could effectively be delivered to HNEpCs. Treatment of HNEpCs with RBD-NPs strongly activated production of several pro-inflammatory cytokines, chemokines, Th1-related cytokines and the monocytes/macrophages growth factors. Interestingly, soluble mediators secreted from RBD-NPs treated HNEpCs significantly upregulated the expression of maturation markers (CD80, CD83, CD86 and HLA-DR) on the MoDCs. CONCLUSION: This study demonstrated that our COVID-19 vaccine candidate drove HNEpCs into immunologically competent cells that not only exerted anti-viral innate immune responses but also potently induced MoDCs maturation.

2.
J Gen Virol ; 103(7)2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35833704

RESUMO

Dengue is a disease that poses a significant global public health concern. Although a tetravalent live-attenuated dengue vaccine has been licensed, its efficacy is still debated due to evidence of vaccine breakthrough infection. To avoid this issue, dengue vaccines should stimulate a high degree of serotype-specific response. Thus, envelope domain III (EDIII), which contains serotype-specific neutralizing epitopes, is an attractive target for dengue vaccine development. In this study, we investigated how EDIII encapsidated in N, N, N-trimethyl chitosan chloride nanoparticles (TMC NPs) stimulates a serotype-specific response and whether this response exerts a potential in vitro breakthrough infection. The immune response to DENV-2 elicited by EDIII TMC NP-immunized mice was monitored. We demonstrated that immunization with EDIII TMC NPs resulted in a high level of anti-EDIII antibody production. These antibodies included IgG, IgG1, and IgG2a subtypes. Importantly, antibodies from the immunized mice exerted efficient neutralizing activity with undetectable antibody dependent enhancement (ADE) activity. We also found that EDIII TMC NPs activated functional EDIII-specific CD4+ and CD8+ T cell responses. In conclusion, EDIII TMC NPs stimulated humoral immunity with a strong neutralizing antibody response, as well as a cellular immune response against DENV-2.


Assuntos
Quitosana , Vacinas contra Dengue , Vírus da Dengue , Dengue , Nanopartículas , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , Anticorpos Facilitadores , Dengue/prevenção & controle , Camundongos , Proteínas do Envelope Viral/genética
3.
Microbiol Immunol ; 64(6): 445-457, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32246487

RESUMO

Chikungunya virus (CHIKV) is a mosquito-borne virus that causes arthralgic fever. Fibroblast-like synoviocytes play a key role in joint damage in inflammatory arthritides and can additionally serve as target cells for CHIKV infection. To gain a better understanding of CHIKV-induced arthralgia, the interaction between CHIKV and synoviocytes was investigated at the protein level. A gel-enhanced liquid chromatography-mass spectrometry (GeLC-MS/MS) approach was used to examine protein expression from primary human fibroblast-like synoviocytes (HFLS) infected with clinical isolates of CHIKV at 12 and 24 hr post infection. Our analysis identified 259 and 241 proteins of known function that were differentially expressed (>1.5 or <-1.5 fold change) following CHIKV infection at 12 and 24 hpi, respectively. These proteins are involved in cellular homeostasis, including cellular trafficking, cytoskeletal organization, immune response, metabolic process, and protein modification. Some of these proteins have previously been reported to participate in arthralgia/arthritis and the death of infected cells. Our results provide information on the CHIKV-induced modulation of cellular proteins of HFLS at an early stage of infection, as well as highlighting biological processes associated with CHIKV infection in the main target cells of the joint.


Assuntos
Febre de Chikungunya , Fibroblastos/imunologia , Interações entre Hospedeiro e Microrganismos/imunologia , Proteoma/imunologia , Sinoviócitos/imunologia , Células Cultivadas , Febre de Chikungunya/imunologia , Febre de Chikungunya/patologia , Febre de Chikungunya/virologia , Vírus Chikungunya/fisiologia , Fibroblastos/patologia , Humanos , Proteômica/métodos , Sinoviócitos/patologia , Replicação Viral
4.
J Gen Virol ; 99(4): 525-535, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29517478

RESUMO

Low-passage clinical isolates of chikungunya virus (CHIKV) were found to be a mixture of large- and small-plaque viruses, with small-plaque viruses being the predominant species. To investigate the contribution of plaque variants to the pathology of the joint, primary human fibroblast-like synoviocytes (HFLS) were used. Large- and small-plaque viruses were purified from two clinical isolates, CHIKV-031C and CHIKV-033C, and were designated CHIKV-031L and CHIKV-031S and CHIKV-033L and CHIKV-033S, respectively. The replication efficiencies of these viruses in HFLSs were compared and it was found that CHIKV-031S and CHIKV-033S replicated with the highest efficiency, while the parental clinical isolates had the lowest efficiency. Interestingly, the cytopathic effects (CPE) induced by these viruses correlated with neither the efficiency of replication nor the plaque size. The small-plaque viruses and the clinical isolates induced cell death rapidly, while large-plaque viruses induced slow CPE in which only 50 % of the cells in infected cultures were rounded up and detached on day 5 of infection. The production of proinflammatory cytokines and chemokines from infected HFLSs was evaluated. The results showed that the large-plaque viruses and the clinical isolates, but not small-plaque variants, were potent inducers of IL-6, IL-8 and MCP-1, and were able to migrate monocytes/macrophages efficiently. Sequencing data revealed a number of differences in amino acid sequences between the small- and large-plaque viruses. The results suggest that it is common for clinical isolates of CHIKV to be heterogeneous, while the variants may have distinct roles in the pathology of the joint.


Assuntos
Febre de Chikungunya/virologia , Vírus Chikungunya/fisiologia , Fibroblastos/virologia , Sinoviócitos/virologia , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/imunologia , Febre de Chikungunya/genética , Febre de Chikungunya/imunologia , Vírus Chikungunya/genética , Vírus Chikungunya/isolamento & purificação , Efeito Citopatogênico Viral , Fibroblastos/imunologia , Humanos , Interleucina-6/genética , Interleucina-6/imunologia , Interleucina-8/genética , Interleucina-8/imunologia , Sinoviócitos/imunologia
5.
Virus Genes ; 54(1): 48-56, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29185115

RESUMO

Chikungunya virus (CHIKV), a mosquito-borne Alphavirus, is the etiological agent of chikungunya fever. CHIKV re-emerged from 2004 onwards, and subsequently caused major outbreaks in many parts of the world including the Indian Ocean islands, Asia, and the Americas. In this study, a large plaque variant of CHIKV isolated from patient in Thailand was subjected to repeated cycles of plaque-purification in Vero cells. The resulting virus produced homogenous large plaques and showed a more pathogenic phenotype than the parental wild-type CHIKV. Whole genome analysis of the large plaque virus in comparison to parental isolate revealed a number of mutations, leading to the following amino acid changes: nsP2 (P618→L), nsP3 (G117→R), and E2 (N187→K). Eight recombinant CHIKVs were constructed to determine which amino acids mediated the large plaque phenotype. The results showed the recombinant virus which contains all three mutations, rCHK-L, produced significantly larger plaques than the other recombinant viruses (p < 0.01). Moreover, the plaque size of the other recombinant virus tended to be smaller if they contained only one or two of the large plaque associated mutations in the viral genome. In conclusion, the combination of all three residues (nsP2-L618, nsP3-R117, and E2-K187) is required to produce the large plaque phenotype of CHIKV.


Assuntos
Vírus Chikungunya/crescimento & desenvolvimento , Vírus Chikungunya/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Fenótipo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Animais , Febre de Chikungunya/virologia , Vírus Chikungunya/isolamento & purificação , Vírus Chikungunya/patogenicidade , Chlorocebus aethiops , Análise Mutacional de DNA , Humanos , Recombinação Genética , Genética Reversa , Inoculações Seriadas , Tailândia , Células Vero , Ensaio de Placa Viral , Virulência , Sequenciamento Completo do Genoma
6.
Asian Pac J Allergy Immunol ; 36(3): 201-205, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28938842

RESUMO

BACKGROUND: Plaque size is a common feature of viral characterization. Small plaque size is used as a marker of attenuation for live-attenuated vaccine development. OBJECTIVE: To investigate whether the naturally occurring plaque size variation reflects virulence of the variants of chikungunya virus (CHIKV). METHODS: We selected and purified a variant with small plaque size from the primary isolate. The viral variant was tested for the plaque morphology, in vitro growth kinetics and mouse neurovirulence in comparison with the parental wild type. RESULTS: The small plaque size variant showed stable homogenous small plaques after 4 plaque purifications. The small plaque virus grew slower and to the lower titer when compared with wild type virus. After 21 days of infection, mice that received small plaque virus showed 98% survival rate while 74% of mice survived after infected with wild type virus. CONCLUSION: The small plaque size variant of CHIKV can be obtained by plaque purification and the virus displays decreased virulence.


Assuntos
Febre de Chikungunya/patologia , Vírus Chikungunya/patogenicidade , Virulência , Replicação Viral , Animais , Camundongos
7.
Biochim Biophys Acta Gen Subj ; 1861(2): 106-111, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27984114

RESUMO

BACKGROUND: Chikungunya fever is an emerging disease caused by the chikungunya virus and is now being spread worldwide by the mosquito Aedes albopictus. The infection can cause a persistent severe joint pain and recent reports link high levels of viremia to neuropathologies and fatalities. The viral protein nsP2 is a multifunctional enzyme that plays several critical roles in virus replication. Virus infection induces oxidative stress in host cells which the virus utilizes to aid viral propagation. Cellular oxidative stress also triggers glutathionylation which is a post-translational protein modification that can modulate physiological roles of affected proteins. METHODS: The nsP2 protease is necessary for processing of the virus nonstructural polyprotein generated during replication. We use the recombinant nsP2 protein to measure protease activity before and after glutathionylation. Mass spectrometry allowed the identification of the glutathione-modified cysteines. Using immunoblots, we show that the glutathionylation of nsP2 occurs in virus-infected cells. RESULTS: We show that in virus-infected cells, the chikungunya nsP2 can be glutathionylated and we show this modification can impact on the protease activity. We also identify 6 cysteine residues that are glutathionylated of the 20 cysteines in the protein. CONCLUSIONS: The virus-induced oxidative stress causes modification of viral proteins which appears to modulate virus protein function. GENERAL SIGNIFICANCE: Viruses generate oxidative stress to regulate and hijack host cell systems and this environment also appears to modulate virus protein function. This may be a general target for intervention in viral pathogenesis.


Assuntos
Vírus Chikungunya/metabolismo , Peptídeo Hidrolases/metabolismo , Proteínas não Estruturais Virais/metabolismo , Linhagem Celular , Febre de Chikungunya/metabolismo , Febre de Chikungunya/virologia , Cisteína/metabolismo , Cisteína Endopeptidases/metabolismo , Glutationa/metabolismo , Células HEK293 , Humanos , Estresse Oxidativo/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Replicação Viral/fisiologia
8.
Microbiol Immunol ; 61(8): 305-317, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28603864

RESUMO

Domain III of E protein of dengue virus (DENV) is a target for vaccine development. Unfortunately, this protein based platform has low general immunogenicity. To circumvent this problem, the use of an adjuvant-nanoparticle delivery system to facilitate immunogenicity of soluble DENV-EDIII protein was investigated. One of the key features of this delivery system is its ability to simultaneously deliver antigens and exert adjuvanticity on specialized immune cells. In this study, N-trimethyl chitosan (TMC) nanoparticles (NPs) were generated to be used as adjuvant and carrier for soluble E-domain III of dengue virus serotype 3 (sEDIII-D3). Using ionotropic gelation, purified sEDIII-D3 was encapsulated into TMC NPs to form EDIII-D3 TMC NPs. After optimization, EDIII-D3 TMC particles exhibited a loading efficiency of 81% and a loading capacity of 41%. The immunogenicity of EDIII-D3 TMC NPs was tested using monocyte-derived dendritic cells (MoDCs). It was found that EDIII-D3 TMC NPs were well taken up by MoDCs. In addition, EDIII-D3 TMC NP treated MoDCs significantly upregulated maturation markers (CD80, CD83, CD86 and HLA-DR) and induced secretion of various cytokines and chemokines (IFN-α, IL-1ß, IL-6, IL-2, IL-12p70, IFN-γ, IL-4, IL-10, IL-8, MCP-1, macrophage inflammatory protein-1ß, granulocyte-colony stimulating factor, granulocyte-macrophage colony-stimulating factor and IL-7). These results indicate that EDIII-D3 TMC NPs are potent immunogens, at least in vitro, with the ability to induce maturation of DCs and highlight the potential use of TMC NPs for enhancing immunogenicity of a non-replicating dengue vaccine.


Assuntos
Células Dendríticas/imunologia , Vacinas contra Dengue/imunologia , Vírus da Dengue/imunologia , Portadores de Fármacos/química , Nanopartículas/química , Proteínas do Envelope Viral/imunologia , Animais , Antígenos Virais/imunologia , Linhagem Celular , Quimiocinas/metabolismo , Quitosana/química , Chlorocebus aethiops , Células Dendríticas/citologia , Vacinas contra Dengue/química , Vírus da Dengue/metabolismo , Humanos , Imunogenicidade da Vacina/imunologia , Células Vero , Proteínas do Envelope Viral/metabolismo
9.
J Gen Virol ; 97(6): 1311-1323, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26935590

RESUMO

Viral adaptability and survival arise due to the presence of quasispecies populations that are able to escape the immune response or produce drug-resistant variants. However, the presence of H5N1 virus with natural mutations acquired without any drug selection pressure poses a great threat. Cloacal samples collected from the 2004-2005 epidemics in Thailand from Asian open-billed storks revealed one major and several minor quasispecies populations with mutations on the oseltamivir (OTV)-binding site of the neuraminidase gene (NA) without prior exposure to a drug. Therefore, this study investigated the binding between the NA-containing novel mutations and OTV drug using molecular dynamic simulations and plaque inhibition assay. The results revealed that the mutant populations, S236F mutant, S236F/C278Y mutant, A250V/V266A/P271H/G285S mutant and C278Y mutant, had a lower binding affinity with OTV as compared with the WT virus due to rearrangement of amino acid residues and increased flexibility in the 150-loop. This result was further emphasized through the IC50 values obtained for the major population and WT virus, 104.74 nM and 18.30 nM, respectively. Taken together, these data suggest that H5N1 viruses isolated from wild birds have already acquired OTV-resistant point mutations without any exposure to a drug.


Assuntos
Antivirais/farmacologia , Farmacorresistência Viral , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Influenza Aviária/virologia , Proteínas Mutantes/genética , Neuraminidase/genética , Oseltamivir/farmacologia , Proteínas Virais/genética , Animais , Aves , Variação Genética , Virus da Influenza A Subtipo H5N1/classificação , Virus da Influenza A Subtipo H5N1/genética , Concentração Inibidora 50 , Testes de Sensibilidade Microbiana , Simulação de Dinâmica Molecular , Proteínas Mutantes/metabolismo , Neuraminidase/metabolismo , Ligação Proteica , Tailândia , Ensaio de Placa Viral , Proteínas Virais/metabolismo
10.
Microb Pathog ; 101: 104-118, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27863885

RESUMO

Over the last decade infections with the mosquito transmitted chikungunya virus (CHIKV) have become a major worldwide concern, and considerable efforts have been made in understanding the interaction of this virus with the host cell machinery. Studies have documented the induction of the unfolded protein response (UPR), as well as the induction of apoptosis and autophagy in response to CHIKV infection. This study comparatively analysed these three processes in two cell lines, Hela and HepG2. Infection of Hela cells was characterized by activation of the PERK/eIF2α branch of the UPR, the induction of autophagy and early apoptosis, while infection of HepG2 cells was characterized by activation of the IRE/XBP1 branch of the UPR, limited or no activation of autophagy and comparatively later apoptosis. These results show that the specific cell context is an important mediator of the host cell response to CHIKV infection.


Assuntos
Vírus Chikungunya/patogenicidade , Estresse do Retículo Endoplasmático , Interações Hospedeiro-Patógeno , Apoptose , Autofagia , Células Epiteliais/fisiologia , Células Epiteliais/virologia , Células HeLa , Células Hep G2 , Hepatócitos/fisiologia , Hepatócitos/virologia , Humanos , Resposta a Proteínas não Dobradas
11.
Virol J ; 13: 142, 2016 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-27538960

RESUMO

BACKGROUND: About half of the world's population are living in the endemic area of dengue viruses implying that a rapid-mass vaccination may be required. In addition, a major target of dengue vaccine are children, thus, a needle-free administration is more attractive. These problems may be overcome by the alternative route of vaccination such as topical, oral and intranasal vaccination. Here, we investigated the possibility to deliver a dengue immunogen intranasally, a painless route of vaccination. The tested immunogen was the domain III of dengue serotype-3 E protein (EDIII-D3) loaded into trimethyl chitosan nanoparticles (EDIII-D3 TMC NPs). The primary human nasal epithelial cells, HNEpCs, were used as an in vitro model for nasal responses. RESULTS: At tested concentrations, EDIII-D3 TMC NPs not only exerted no detectable toxicity toward HNEpC cultures but also efficiently delivered EDIII-D3 immunogens into HNEpCs. Moreover, HNEpCs quickly and strongly produced proinflammatory cytokines (IL-1ß, IL-6, TNF-α), type-I IFN, the growth factors (GM-CSF, IL-7), the chemokines (MCP-1, MIP-1ß, IL-8), Th1-related cytokines (IL-2, IL-12p70, IL-17, IFN-γ) and Th2-related cytokine (IL-4) in response to EDIII-D3 TMC NPs treatment. CONCLUSIONS: A potential mucosal delivery system for dengue immunogens was revealed and found to stimulate a strong local innate antiviral response which possibly leading to a systemic adaptive immunity.


Assuntos
Vacinas contra Dengue/imunologia , Vírus da Dengue/imunologia , Dengue/virologia , Células Epiteliais/imunologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Administração Intranasal , Dengue/imunologia , Vacinas contra Dengue/administração & dosagem , Vacinas contra Dengue/genética , Vírus da Dengue/genética , Células Epiteliais/virologia , Humanos , Interleucina-2/imunologia , Interleucina-8/imunologia , Nariz/citologia , Nariz/imunologia , Nariz/virologia , Células Th1/imunologia , Células Th2/imunologia , Vacinação , Proteínas do Envelope Viral/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
12.
J Med Virol ; 87(5): 733-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25656089

RESUMO

In addition to fever, rash, and arthralgia/arthritis, myalgia is another dominant symptom in Chikungunya virus (CHIKV) infection. How CHIKV induces myalgia is unclear. To better understand the viral factors involved in CHIKV-induced myalgia, CHIKVs were isolated from patients with and without myalgia designated myalgia-CHIKV and mild-CHIKV, respectively. The response of myoblasts to infection by the two groups of clinical isolates of CHIKV was investigated. Both groups of CHIKV replicated well in primary human myoblasts. However, the myalgia-CHIKVs replicated to a higher titer and caused the death of infected myoblast more rapidly than the mild-CHIKVs. CHIKV-infected myoblasts increased production of four out of five inflammatory cytokines examined (MCP-1, IP-10, MIP-1α, and IL-8) in comparison to mock-infected cells. Comparison between the myoblast inflammatory cytokine responses showed that myalgia-CHIKVs were stronger activators of cytokines than mild-CHIKVs. This means that recent epidemic strains of CHIKV exhibited different degrees of myoblast permissiveness as evidenced by differences in the ability to replicate and to stimulate inflammatory responses in myoblasts. This data suggest that the myopathic syndrome in recent epidemics is dependent upon the strain of CHIKV.


Assuntos
Febre de Chikungunya/virologia , Vírus Chikungunya/fisiologia , Mialgia/virologia , Mioblastos/imunologia , Mioblastos/virologia , Replicação Viral , Adulto , Células Cultivadas , Febre de Chikungunya/epidemiologia , Febre de Chikungunya/patologia , Vírus Chikungunya/crescimento & desenvolvimento , Vírus Chikungunya/isolamento & purificação , Citocinas/metabolismo , Humanos , Carga Viral
13.
Microbiol Immunol ; 59(3): 129-41, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25643977

RESUMO

Chikungunya virus (CHIKV) is a re-emerging mosquito-borne alphavirus that recently caused large epidemics in islands in, and countries around, the Indian Ocean. There is currently no specific drug for therapeutic treatment or for use as a prophylactic agent against infection and no commercially available vaccine. Prohibitin has been identified as a receptor protein used by chikungunya virus to enter mammalian cells. Recently, synthetic sulfonyl amidines and flavaglines (FLs), a class of naturally occurring plant compounds with potent anti-cancer and cytoprotective and neuroprotective activities, have been shown to interact directly with prohibitin. This study therefore sought to determine whether three prohibitin ligands (sulfonyl amidine 1 m and the flavaglines FL3 and FL23) were able to inhibit CHIKV infection of mammalian Hek293T/17 cells. All three compounds inhibited infection and reduced virus production when cells were treated before infection but not when added after infection. Pretreatment of cells for only 15 minutes prior to infection followed by washing out of the compound resulted in significant inhibition of entry and virus production. These results suggest that further investigation of prohibitin ligands as potential Chikungunya virus entry inhibitors is warranted.


Assuntos
Antivirais/farmacologia , Benzofuranos/farmacologia , Febre de Chikungunya/virologia , Vírus Chikungunya/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Antivirais/síntese química , Benzofuranos/síntese química , Vírus Chikungunya/fisiologia , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Humanos , Replicação Viral/efeitos dos fármacos
14.
J Transl Med ; 12: 96, 2014 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-24721947

RESUMO

BACKGROUND: Chikungunya fever (CHIKF) is a recently re-emerged mosquito transmitted viral disease caused by the chikungunya virus (CHIKV), an Alphavirus belonging to the family Togaviridae. Infection of humans with CHIKV can result in CHIKF of variable severity, although the factors mediating disease severity remain poorly defined. METHODS: White blood cells were isolated from blood samples collected during the 2009-2010 CHIKF outbreak in Thailand. Clinical presentation and viral load data were used to classify samples into three groups, namely non chikungunya fever (non-CHIKF), mild CHIKF, and severe CHIKF. Five samples from each group were analyzed for protein expression by GeLC-MS/MS. RESULTS: CHIKV proteins (structural and non-structural) were found only in CHIKF samples. A total of 3505 human proteins were identified, with 68 proteins only present in non-CHIKF samples. A total of 240 proteins were found only in CHIKF samples, of which 65 and 46 were found only in mild and severe CHIKF samples respectively. Proteins with altered expression mapped predominantly to cellular signaling pathways (including toll-like receptor and PI3K-Akt signaling) although many other processes showed altered expression as a result of CHIKV infection. Expression of proteins consistent with the activation of the inflammasome was detected, and quantitation of (pro)-caspase 1 at the protein and RNA levels showed an association with disease severity. CONCLUSIONS: This study confirms the infection of at least a component of white blood cells by CHIKV, and shows that CHIKV infection results in activation of the inflammasome in a manner that is associated with disease severity.


Assuntos
Febre de Chikungunya/sangue , Linfócitos/metabolismo , Proteômica , Sequência de Bases , Cromatografia Líquida , Primers do DNA , Humanos , Reação em Cadeia da Polimerase em Tempo Real , Espectrometria de Massas em Tandem
15.
Arch Virol ; 159(12): 3353-64, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25168043

RESUMO

Chikungunya virus (CHIKV), the virus responsible for the disease chikungunya fever in humans, is transmitted by Aedes mosquitoes. While significant progress has been made in understanding the process by which CHIKV enters into mammalian cells, far less progress has been made in understanding the CHIKV entry process in insect cells. This study sought to identify mosquito-cell-expressed CHIKV-binding proteins through a combination of virus overlay protein binding assays (VOPBA) and mass spectroscopy. A 50-kDa CHIKV-binding protein was identified as the ATP synthase ß subunit (ATPSß). Co-immunoprecipitation studies confirmed the interaction, and colocalization analysis showed cell-surface and intracellular co-localization between CHIKV and ATPSß. Both antibody inhibition and siRNA-mediated downregulation experiments targeted to ATPSß showed a significant reduction in viral entry and virus production. These results suggest that ATPSß is a CHIKV-binding protein capable of mediating the entry of CHIKV into insect cells.


Assuntos
Aedes/virologia , Vírus Chikungunya/fisiologia , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Receptores Virais/metabolismo , Internalização do Vírus , Animais , Expressão Gênica , Interações Hospedeiro-Patógeno , Espectrometria de Massas , Ligação Proteica , Ligação Viral
16.
Carbohydr Polym ; 337: 122132, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38710548

RESUMO

N,N,N-Trimethyl chitosan (TMC) is a quaternized chitosan with versatile biological features. However, low mechanical strength limits its uses, for example, as hydrogels for tissue engineering applications. This study illustrates a viable synthesis of metal/polymer hybrid, core-shell colloidal particles and their use as reinforcing and antioxidant fillers for TMC hydrogels. The core-shell particles were initially synthesized by surfactant-free emulsion polymerization, induced by a photo-redox initiating system of riboflavin assisted by a 3° amine and 2° alcohol co-initiators. The synthesized core-shell particles were based on two polymeric shells: TMC and chitosan, and two polymeric cores: poly (hydroxypropyl methacrylate) (PHPMA) and poly(2-hydroxy ethyl methacrylate) (PHEMA). The presence of both 3° amine on TMC and 2° alcohol on HPMA monomer enhanced the photopolymerization performance. The TMC-based particles had sizes of 122-154 nm and zeta potentials of 10-35 mV, bringing the colloidal stability in the 4-10 pH range. Furthermore, due to the presence of TMC on the shell layer, the core-shell particles could be used as templates to grow the Ag/Au bimetallic nanoparticles with alloy and core-shell types through a thermal reduction. The prepared hybrid particles were incorporated in TMC hydrogels as a multifunctional filler, improving their mechanical and antioxidant properties.

17.
Vaccine ; 42(17): 3664-3673, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38714446

RESUMO

With the continuous emergence of coronavirus disease 2019 (COVID-19) waves, the scientific community has developed a vaccine that offers broad-spectrum protection at virus-targeted organs for inhibiting the transmission and protection of disease development. In the present study, a bivalent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine containing receptor-binding domain (RBD) protein of spike from Wuhan-1 and omicron BA.1 loaded in nanoparticles, bivalent RBD NPs, was developed. The immunogenicity and protective efficacy of this vaccine candidate were evaluated using an in vivo model. Results showed that mice that received intranasal cGAMP-adjuvanted bivalent RBD-NPs vaccine elicited robust and durable antibody responses. The stimulated antibody broadly neutralized the ancestral strain and variants of concerns (delta and omicron BA.1) in the upper and lower respiratory tracts. Furthermore, the immunized mice developed T-cell response in their lung tissue. Importantly, intranasal immunization with this vaccine candidate efficiently protected mice from nasal infection caused by both Wuhan-1 and BA.1 viruses. Immunized mice that remained susceptible to nasal infection did not develop any symptoms. This is because activated responses in the nasal cavity significantly suppressed virus production. Another word is this nasal vaccine completely protected the mice from disease development and mortality. Therefore, the bivalent RBD vaccine platform has potential to be developed into an anti-SARS-CoV-2 universal vaccine.


Assuntos
Administração Intranasal , Anticorpos Neutralizantes , Anticorpos Antivirais , Vacinas contra COVID-19 , COVID-19 , Camundongos Endogâmicos BALB C , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Animais , Vacinas contra COVID-19/imunologia , Vacinas contra COVID-19/administração & dosagem , Camundongos , COVID-19/prevenção & controle , COVID-19/imunologia , SARS-CoV-2/imunologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Glicoproteína da Espícula de Coronavírus/imunologia , Feminino , Modelos Animais de Doenças , Pulmão/virologia , Pulmão/imunologia , Pulmão/patologia , Nanopartículas/administração & dosagem , Imunização
18.
Vaccine ; 41(9): 1638-1648, 2023 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-36740559

RESUMO

Dengue is the most prevalent mosquito-borne viral disease and continues to be a global public health concern. Although a licensed dengue vaccine is available, its efficacy and safety profile are not satisfactory. Hence, there remains a need for a safe and effective dengue vaccine. We are currently developing a bivalent dengue vaccine candidate. This vaccine candidate is composed of a C-terminus truncated non-structural protein 1 (NS11-279) and envelope domain III (EDIII) of DENV-2 encapsidated in the nanocarriers, N, N, N-trimethyl chitosan nanoparticles (TMC NPs). The immunogenicity of this bivalent vaccine candidate was investigated in the present study using BALB/c mice. In this work, we demonstrate that NS1 + EDIII TMC NP-immunized mice strongly elicited antigen-specific antibody responses (anti-NS1 and anti-EDIII IgG) and T-cell responses (NS1- and EDIII-specific-CD4+ and CD8+ T cells). Importantly, the antibody response induced by NS1 + EDIII TMC NPs provided antiviral activities against DENV-2, including serotype-specific neutralization and antibody-mediated complement-dependent cytotoxicity. Moreover, the significant upregulation of Th1- and Th2-associated cytokines, as well as the increased levels of antigen-specific IgG2a and IgG1, indicated a balanced Th1/Th2 response. Collectively, our findings suggest that NS1 + EDIII TMC NPs induced protective responses that can not only neutralize infectious DENV-2 but also eliminate DENV-2-infected cells.


Assuntos
Vacinas contra Dengue , Vírus da Dengue , Dengue , Nanopartículas , Animais , Camundongos , Dengue/prevenção & controle , Anticorpos Antivirais , Linfócitos T CD8-Positivos , Proteínas do Envelope Viral
19.
Vaccines (Basel) ; 11(8)2023 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-37631912

RESUMO

The uneven immunogenicity of the attenuated tetravalent dengue vaccine has made it difficult to achieve balanced protection against all four serotypes of the dengue virus (DENV). To overcome this problem, non-replicative vaccines have come into focus, as their immunogenicity is adjustable. This approach is excellent for multivalent vaccines but commonly faces the issue of low immunogenicity. In this present study, we developed a non-replicating dengue vaccine composed of UV-inactivated dengue virus-2 (UV-DENV-2) and DENV-2 NS1-279 protein encapsidated within nanoparticles. This vaccine candidate was administered in the presence of BCG cell wall cytoskeleton (BCG-CWS) as an adjuvant. We revealed, here, that encapsidated immunogens with BCG-CWS exerted potent activities on both B and T cells and elicited Th-1/Th-2 responses in mice. This was evidenced by BCG-CWS significantly augmenting antibody-mediated complement-fixing activity, strongly stimulating the antigen-specific polyfunctional T cell responses, and activating mixed Th-1/Th-2 responses specific to DENV-2- and NS1-279 antigens. In conclusion, BCG-CWS potently adjuvanted the inactivated DENV-2 and DENV subunit immunogens. The mechanism of adjuvanticity remains unclear. This study revealed the potential use of BCG-CWS in vaccine development.

20.
Viruses ; 15(10)2023 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-37896789

RESUMO

Every year, dengue virus (DENV) affects millions of people. Currently, there are no approved drugs for the treatment of DENV infection. Autophagy is a conserved degradation process that was shown to be induced by DENV infection and required for optimal DENV replication. The modulation of autophagy is, therefore, considered an attractive target to treat DENV infection. This study carried out a high-content image screen analysis using Crispr-Cas9 GFP-LC3 knocked-in HeLa cells of a compound library synthesized from or inspired by natural products and their biocongener precursors to discover novel autophagy inhibitors. The screen identified Ka-003 as the most effective compound for decreasing the number of autophagic vacuoles inside cells upon autophagy induction. Ka-003 could inhibit autophagy in a dose-dependent manner at low micromolar concentrations. More importantly, Ka-003 demonstrated the concentration-dependent inhibition of DENV production in Crispr-Cas9 GFP-LC3 knocked-in THP-1 monocytes. The core structure of Ka-003, which is a methyl cyclohexene derivative, resembles those found in mulberry plants, and could be synthetically prepared in a bioinspired fashion. Taken together, data indicate that Ka-003 hampered autophagy and limited DENV replication. The low cytotoxicity of Ka-003 suggests its therapeutic potential, which warrants further studies for the lead optimization of the compound for dengue treatment.


Assuntos
Vírus da Dengue , Dengue , Humanos , Vírus da Dengue/fisiologia , Células HeLa , Autofagia/fisiologia , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA