Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Virol Sin ; 38(5): 787-800, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37423308

RESUMO

The weakened protective efficacy of COVID-19 vaccines and antibodies caused by SARS-CoV-2 variants presents a global health emergency, which underscores the urgent need for universal therapeutic antibody intervention for clinical patients. Here, we screened three alpacas-derived nanobodies (Nbs) with neutralizing activity from twenty RBD-specific Nbs. The three Nbs were fused with the Fc domain of human IgG, namely aVHH-11-Fc, aVHH-13-Fc and aVHH-14-Fc, which could specifically bind RBD protein and competitively inhibit the binding of ACE2 receptor to RBD. They effectively neutralized SARS-CoV-2 pseudoviruses D614G, Alpha, Beta, Gamma, Delta, and Omicron sub-lineages BA.1, BA.2, BA.4, and BA.5 and authentic SARS-CoV-2 prototype, Delta, and Omicron BA.1, BA.2 strains. In mice-adapted COVID-19 severe model, intranasal administration of aVHH-11-Fc, aVHH-13-Fc and aVHH-14-Fc effectively protected mice from lethal challenges and reduced viral loads in both the upper and lower respiratory tracts. In the COVID-19 mild model, aVHH-13-Fc, which represents the optimal neutralizing activity among the above three Nbs, effectively protected hamsters from the challenge of SARS-CoV-2 prototype, Delta, Omicron BA.1 and BA.2 by significantly reducing viral replication and pathological alterations in the lungs. In structural modeling of aVHH-13 and RBD, aVHH-13 binds to the receptor-binding motif region of RBD and interacts with some highly conserved epitopes. Taken together, our study illustrated that alpaca-derived Nbs offered a therapeutic countermeasure against SARS-CoV-2, including those Delta and Omicron variants which have evolved into global pandemic strains.


Assuntos
COVID-19 , Camelídeos Americanos , Anticorpos de Domínio Único , Cricetinae , Humanos , Animais , Camundongos , COVID-19/terapia , SARS-CoV-2/genética , Vacinas contra COVID-19 , Anticorpos de Domínio Único/genética , Modelos Animais de Doenças , Imunoglobulina G , Anticorpos Neutralizantes , Anticorpos Antivirais/uso terapêutico , Glicoproteína da Espícula de Coronavírus/genética
2.
Front Immunol ; 14: 1066730, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36875106

RESUMO

The emergence of SARS-CoV-2 variants stresses the continued need for broad-spectrum therapeutic antibodies. Several therapeutic monoclonal antibodies or cocktails have been introduced for clinical use. However, unremitting emerging SARS-CoV-2 variants showed reduced neutralizing efficacy by vaccine induced polyclonal antibodies or therapeutic monoclonal antibodies. In our study, polyclonal antibodies and F(ab')2 fragments with strong affinity produced after equine immunization with RBD proteins produced strong affinity. Notably, specific equine IgG and F(ab')2 have broad and high neutralizing activity against parental virus, all SARS-CoV-2 variants of concern (VOCs), including B.1.1,7, B.1.351, B.1.617.2, P.1, B.1.1.529 and BA.2, and all variants of interest (VOIs) including B.1.429, P.2, B.1.525, P.3, B.1.526, B.1.617.1, C.37 and B.1.621. Although some variants weaken the neutralizing ability of equine IgG and F(ab')2 fragments, they still exhibited superior neutralization ability against mutants compared to some reported monoclonal antibodies. Furthermore, we tested the pre-exposure and post-exposure protective efficacy of the equine immunoglobulin IgG and F(ab')2 fragments in lethal mouse and susceptible golden hamster models. Equine immunoglobulin IgG and F(ab')2 fragments effectively neutralized SARS-CoV-2 in vitro, fully protected BALB/c mice from the lethal challenge, and reduced golden hamster's lung pathological change. Therefore, equine pAbs are an adequate, broad coverage, affordable and scalable potential clinical immunotherapy for COVID-19, particularly for SARS-CoV-2 VOCs or VOIs.


Assuntos
COVID-19 , SARS-CoV-2 , Cricetinae , Animais , Cavalos , Humanos , Camundongos , Roedores , Mesocricetus , Anticorpos Monoclonais , Anticorpos Amplamente Neutralizantes , Imunoglobulina G , Camundongos Endogâmicos BALB C
3.
Emerg Microbes Infect ; 12(1): 2149351, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36453198

RESUMO

Marburg virus disease (MVD) is a lethal viral haemorrhagic fever caused by Marburg virus (MARV) with a case fatality rate as high as 88%. There is currently no vaccine or antiviral therapy approved for MVD. Due to high variation among MARV isolates, vaccines developed against one strain fail to protect against other strains. Here we report that three recombinant rabies virus (RABV) vector vaccines encoding two copies of GPs covering both MARV lineages induced pseudovirus neutralizing antibodies in BALB/c mice. Furthermore, high-affinity human neutralizing antibodies were isolated from a humanized mouse model. The three vaccines produced a Th1-biased serological response similar to that of human patients. Adequate sequential immunization enhanced the production of neutralizing antibodies. Virtual docking suggested that neutralizing antibodies induced by the Angola strain seemed to be able to hydrogen bond to the receptor-binding site (RBS) in the GP of the Ravn strain through hypervariable regions 2 (CDR2) and CDR3 of the VH region. These findings demonstrate that three inactivated vaccines are promising candidates against different strains of MARV, and a novel fully humanized neutralizing antibody against MARV was isolated.


Assuntos
Doença do Vírus de Marburg , Marburgvirus , Vírus da Raiva , Vacinas Virais , Humanos , Animais , Camundongos , Marburgvirus/genética , Anticorpos Neutralizantes , Vírus da Raiva/genética , Anticorpos Antivirais , Glicoproteínas , Doença do Vírus de Marburg/prevenção & controle , Vacinas Virais/genética
4.
Front Immunol ; 13: 992787, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36211410

RESUMO

The coronavirus disease 2019 pandemic has caused more than 532 million infections and 6.3 million deaths to date. The reactive and neutralizing fully human antibodies of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are effective detection tools and therapeutic measures. During SARS-CoV-2 infection, a large number of SARS-CoV-2 reactive and neutralizing antibodies will be produced. Most SARS-CoV-2 reactive and neutralizing fully human antibodies are isolated from human and frequently encoded by convergent heavy-chain variable genes. However, SARS-CoV-2 viruses can mutate rapidly during replication and the resistant variants of neutralizing antibodies easily survive and evade the immune response, especially in the face of such focused antibody responses in humans. Therefore, additional tools are needed to develop different kinds of fully human antibodies to compensate for current deficiency. In this study, we utilized antibody humanized CAMouseHG mice to develop a rapid antibody discovery method and examine the antibody repertoire of SARS-CoV-2 RBD-reactive hybridoma cells derived from CAMouseHG mice by using high-throughput single-cell V(D)J sequencing analysis. CAMouseHG mice were immunized by 28-day rapid immunization method. After electrofusion and semi-solid medium screening on day 12 post-electrofusion, 171 hybridoma clones were generated based on the results of SARS-CoV-2 RBD binding activity assay. A rather obvious preferential usage of IGHV6-1 family was found in these hybridoma clones derived from CAMouseHG mice, which was significantly different from the antibodies found in patients with COVID-19. After further virus neutralization screening and antibody competition assays, we generated a noncompeting two-antibody cocktail, which showed a potent prophylactic protective efficacy against SARS-CoV-2 in cynomolgus macaques. These results indicate that humanized CAMouseHG mice not only provide a valuable platform to obtain fully human reactive and neutralizing antibodies but also have a different antibody repertoire from humans. Thus, humanized CAMouseHG mice can be used as a good complementary tool in discovery of fully human therapeutic and diagnostic antibodies.


Assuntos
COVID-19 , SARS-CoV-2 , Animais , Anticorpos Monoclonais , Anticorpos Neutralizantes , Anticorpos Antivirais , Humanos , Hibridomas/metabolismo , Camundongos , Glicoproteína da Espícula de Coronavírus
5.
Front Microbiol ; 13: 927122, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35756049

RESUMO

Marburg virus (MARV) is one of the principal members of the filovirus family, which can cause fatal hemorrhagic fever in humans. There are currently no prophylactic and therapeutic drugs on the market, and the high pathogenicity and infectivity of MARV make its research highly dependent on biosafety level 4 conditions, severely hindering the development of vaccines and therapies. Therefore, the development of medicines, such as MARV serological diagnosis, vaccines, and therapeutic antibody drugs, urgently needs a safe, convenient, and biosafety level 2 detection method to measure the neutralizing activity of MARV antibodies. To this end, we report a neutralization assay relying on a Rabies virus (RABV) reverse genetic operating system. We constructed infectious clones carrying the eGFP reporter gene and the full length of the original unmodified MARV GP gene. Based on the critical parameters of phylogenetic analysis, recombinant viruses targeting representative strains in the two major MARV lineages were successfully rescued. These pseudoviruses are safe in mice, and their inability to infect cells after being neutralized by antibodies can be visualized under a fluorescence microscope. We tested the system using the neutralizing antibody MR191. MR191 can significantly block the infection of BSR cells with pseudovirus. We compared it with the traditional lentivirus-type pseudovirus system to verify the system's credibility and obtained the same results as reported in the literature. In general, we have established a safe and visualized method for evaluating the neutralizing activity of MARV antibodies. Compared with traditional methods, it has the advantages of convenient operation, short cycle, and low cost. It is a candidate method that can replace actual viruses for a neutralization assay.

6.
Front Immunol ; 13: 823949, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35173733

RESUMO

Middle East respiratory syndrome coronavirus (MERS-CoV) is an emergent coronavirus that has caused frequent zoonotic events through camel-to-human spillover. An effective camelid vaccination strategy is probably the best way to reduce human exposure risk. Here, we constructed and evaluated an inactivated rabies virus-vectored MERS-CoV vaccine in mice, camels, and alpacas. Potent antigen-specific antibody and CD8+ T-cell responses were generated in mice; moreover, the vaccination reduced viral replication and accelerated virus clearance in MERS-CoV-infected mice. Besides, protective antibody responses against both MERS-CoV and rabies virus were induced in camels and alpacas. Satisfyingly, the immune sera showed broad cross-neutralizing activity against the three main MERS-CoV clades. For further characterization of the antibody response induced in camelids, MERS-CoV-specific variable domains of heavy-chain-only antibody (VHHs) were isolated from immunized alpacas and showed potent prophylactic and therapeutic efficacies in the Ad5-hDPP4-transduced mouse model. These results highlight the inactivated rabies virus-vectored MERS-CoV vaccine as a promising camelid candidate vaccine.


Assuntos
Camelídeos Americanos/virologia , Camelus/virologia , Infecções por Coronavirus/veterinária , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Linfócitos T CD8-Positivos/imunologia , Camelídeos Americanos/imunologia , Camelus/imunologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Cricetinae , Feminino , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vírus da Raiva/genética , Vírus da Raiva/imunologia , Vacinação , Vacinas Sintéticas/imunologia , Células Vero , Vacinas Virais/genética
7.
PLoS Negl Trop Dis ; 15(3): e0009227, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33647020

RESUMO

Since its first emergence in 2012, cases of infection with Middle East respiratory syndrome coronavirus (MERS-CoV) have continued to occur. At the end of January 2020, 2519 laboratory confirmed cases with a case-fatality rate of 34.3% have been reported. Approximately 84% of human cases have been reported in the tropical region of Saudi Arabia. The emergence of MERS-CoV has highlighted need for a rapid and accurate assay to triage patients with a suspected infection in a timely manner because of the lack of an approved vaccine or an effective treatment for MERS-CoV to prevent and control potential outbreaks. In this study, we present two rapid and visual nucleic acid assays that target the MERS-CoV UpE and N genes as a panel that combines reverse transcription recombinase polymerase amplification with a closed vertical flow visualization strip (RT-RPA-VF). This test panel was designed to improve the diagnostic accuracy through dual-target screening after referencing laboratory testing guidance for MERS-CoV. The limit of detection was 1.2×101 copies/µl viral RNA for the UpE assay and 1.2 copies/µl viral RNA for the N assay, with almost consistent with the sensitivity of the RT-qPCR assays. The two assays exhibited no cross-reactivity with multiple CoVs, including the bat severe acute respiratory syndrome related coronavirus (SARSr-CoV), the bat coronavirus HKU4, and the human coronaviruses 229E, OC43, HKU1 and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Furthermore, the panel does not require sophisticated equipment and provides rapid detection within 30 min. This panel displays good sensitivity and specificity and may be useful to rapidly detect MERS-CoV early during an outbreak and for disease surveillance.


Assuntos
Técnicas de Laboratório Clínico/métodos , Infecções por Coronavirus/virologia , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Técnicas de Diagnóstico Molecular/métodos , Infecções por Coronavirus/diagnóstico , Infecções por Coronavirus/epidemiologia , Humanos , RNA Viral/genética , Reação em Cadeia da Polimerase em Tempo Real/métodos , Transcrição Reversa , Arábia Saudita/epidemiologia , Sensibilidade e Especificidade , Proteínas não Estruturais Virais/genética
8.
Front Microbiol ; 11: 590732, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33281787

RESUMO

Rift Valley fever (RVF) is a severe infectious disease, which can through mosquito bites, direct contact and aerosol transmission infect sheep, goats, people, camels, cattle, buffaloes, and so on. In this paper, a conserved region of the S RNA segment of Rift Valley fever virus (RVFV) ZH501 strain was used as target sequence. The RVFV RT-LAMP-VF assay was successfully established combined reverse transcription-loop-mediated isothermal amplification with a vertical flow visualization strip. The detection limit is up to 1.94 × 100 copies/µl of synthesized RVFV-RNA. RNA extracted from cell culture of an inactivated RVFV-BJ01 strain was also used as templates, and the detection limit is 1.83 × 103 copies/µl. In addition, there was no cross-reactivity with other viruses that can cause similar fever symptoms. The RVFV-LAMP-VF assay exhibited very high levels of diagnostic sensitivity, which had 100-fold more sensitive than RVFV real-time RT-PCR assay. Accordingly, the RVFV RT-LAMP-VF assay developed in this study is suitable for the rapid and sensitive diagnosis of RVFV without specialized equipment and can rapidly complete detection within 60 min, and the results are visible by vertical flow visualization strip within 5 min.

9.
Viruses ; 11(10)2019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31597372

RESUMO

Rift Valley fever virus (RVFV), which causes Rift Valley fever (RVF), is a mosquito-borne zoonotic pathogen that causes serious morbidity and mortality in livestock and humans. RVF is a World Health Organization (WHO) priority disease and, together with rabies, is a major health burden in Africa. Here, we present the development and characterization of an inactivated recombinant RVFV and rabies virus (RABV) vaccine candidate (rSRV9-eGn). Immunization with rSRV9-eGn stimulated the production of RVFV-specific IgG antibodies and induced humoral and cellular immunity in mice but did not induce the production of neutralizing antibodies. IgG1 and IgG2a were the main isotypes observed by IgG subtype detection, and IgG3 antibodies were not detected. The ratios of IgG1/IgG2a > 1 indicated a Type 2 humoral immune response. An effective vaccine is intended to establish a long-lived population of memory T cells, and mice generated memory cells among the proliferating T cell population after immunization with rSRV9-eGn, with effector memory T cells (TEM) as the major population. Due to the lack of prophylactic treatment experiments, it is impossible to predict whether this vaccine can protect animals from RVFV infection with only high titres of anti-RVFV IgG antibodies and no neutralizing antibodies induced, and thus, protection confirmation needs further verification. However, this RVFV vaccine designed with RABV as the vector provides ideas for the development of vaccines that prevent RVFV and RABV infections.


Assuntos
Vacina Antirrábica/genética , Vacina Antirrábica/imunologia , Vírus da Raiva/genética , Febre do Vale de Rift/imunologia , Febre do Vale de Rift/prevenção & controle , Vírus da Febre do Vale do Rift/imunologia , Vacinas Virais/imunologia , Adjuvantes Imunológicos , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Modelos Animais de Doenças , Feminino , Imunidade Humoral/imunologia , Imunização , Imunoglobulina G , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T/imunologia , Vacinação , Vacinas de Produtos Inativados/imunologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Replicação Viral
10.
Virus Res ; 270: 197638, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31173772

RESUMO

Adjuvants are important components of vaccination strategies because they boost and accelerate the immune response. The aim of this study was to investigate the adjuvant activity of PCP-II, a polysaccharide isolated from Poria cocos, together with an inactivated rabies vaccine. The polysaccharide PCP-II was compared with the common veterinary rabies vaccine adjuvant Alhydrogel by co-administration of either adjuvant with the inactivated rabies virus rCVS-11-G to mice via the intramuscular route. Blood samples were collected to determine the virus-neutralizing antibody (VNA) titer and assess activation of B and T lymphocytes. Inguinal lymph node samples were collected, and proliferation of B lymphocytes was measured. Splenocytes were isolated, and antigen-specific cellular immune responses were evaluated by enzyme-linked immunospot and immunosorbent assays (ELISpot assay and ELISA, respectively). The results showed that PCP-II enhanced and promoted an increase in the VNA titer in the mice compared to Alhydrogel. Flow cytometry assays revealed that the polysaccharide activated more B lymphocytes in the lymph nodes and more B and T lymphocytes in the blood. Assessment of antigen-specific cellular immune responses showed that PCP-II strongly induced T lymphocyte proliferation in the spleen and high levels of cytokine secretion from splenocytes. All of these data suggest that PCP-II possesses excellent adjuvant activity and enhances both cellular and humoral immunity in mice. After examining the adjuvant activities of PCP-II in mice, dogs were immunized with rCVS-11-G together with Alhydrogel or PCP-II as an adjuvant; the control group was injected with a commercial rabies vaccine. Serum samples were collected, and the VNA titers were measured. PCP-II caused increases in the VNA titers in both the booster and single-dose immunization tests when co-administered with rCVS-11-G compared with Alhydrogel. The VNA titer of the commercial vaccine group was also significantly lower than that of the PCP-II group. These data indicate that PCP-II is an excellent candidate adjuvant for inactive rabies vaccines in the veterinary setting.


Assuntos
Adjuvantes Imunológicos/química , Anticorpos Antivirais/sangue , Polissacarídeos Fúngicos/imunologia , Vacina Antirrábica/imunologia , Raiva/prevenção & controle , Wolfiporia/química , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos Neutralizantes/sangue , Citocinas/imunologia , Cães , Feminino , Polissacarídeos Fúngicos/administração & dosagem , Imunidade Celular , Camundongos , Camundongos Endogâmicos BALB C , Vacina Antirrábica/administração & dosagem , Vírus da Raiva , Vacinação , Vacinas de Produtos Inativados/imunologia
11.
Virus Genes ; 55(1): 95-103, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30519855

RESUMO

Feline panleukopenia virus (FPV) infects cats and can be fatal to kittens. There is evidence that canine parvovirus originated from FPV, which makes FPV important in studies of the family Parvoviridae. In the present study, the entire genome of FPV strain HH-1/86 was converted into a full-length infectious clone (pFPV). The FPV strain HH-1/86 has a 5123-nt single stranded DNA genome with a Y-shaped inverted 3' terminal repeat (ITR) and a U-shaped inverted 5' ITR. Feline kidney cells (F81) were transfected with the pFPV clone which contained a genetic marker, and a rescued virus was obtained (rFPV). The rFPV was identified by its cytopathic effects, indirect immunofluorescence, growth curve analysis, western blot assay and hemagglutination, and was indistinguishable from the parent virus. The FPV infectious clone will facilitate the study of pathogenicity and viral replication of FPV and the inter-species transmission of parvoviruses.


Assuntos
Vírus da Panleucopenia Felina/genética , Panleucopenia Felina/virologia , Genética Reversa , Animais , Gatos , Clonagem Molecular , DNA Viral , Marcadores Genéticos , Genoma Viral , Genômica/métodos , Hemaglutinação , Hemaglutininas Virais/metabolismo , Genética Reversa/métodos , Sequenciamento Completo do Genoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...