Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 233
Filtrar
1.
Antiviral Res ; 227: 105905, 2024 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-38740191

RESUMO

The rapid emergence of Severe Acute Respiratory Syndrome Coronavirus type 2 (SARS-CoV-2) variants, coupled with severe immune evasion and imprinting, has jeopardized the vaccine efficacy, necessitating urgent development of broad protective vaccines. Here, we propose a strategy employing recombinant rabies viruses (RABV) to create a universal SARS-CoV-2 vaccine expressing heterologous tandem receptor-binding domain (RBD) trimer from the SARS-CoV-2 Prototype, Delta, and Omicron strains (SRV-PDO). The results of mouse immunization indicated that SRV-PDO effectively induced cellular and humoral immune responses, and demonstrated higher immunogenicity and broader SARS-CoV-2 neutralization compared to the recombinant RABVs that only expressed RBD monomers. Moreover, SRV-PDO exhibited full protection against SARS-CoV-2 in the challenge assay. This study demonstrates that recombinant RABV expressing tandem RBD-heterotrimer as a multivalent immunogen could elicit a broad-spectrum immune response and potent protection against SARS-CoV-2, making it a promising candidate for future human or veterinary vaccines and offering a novel perspective in other vaccine design.

2.
Poult Sci ; 103(7): 103800, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38743966

RESUMO

The combination of inflammatory factors resulting from an influenza A virus infection is one of the main causes of death in host animals. Studies have shown that guinea pig guanosine monophosphate binding protein 1 (guanylate-binding protein 1, gGBP1) can downregulate cytokine production induced by the influenza virus. Therefore, exploring the innate immune defense mechanism of GBP1 in the process of H5N1 influenza virus infection has important implications for understanding the pathogenic mechanism, disease prevention, and the control of influenza A virus infections. We found that, in addition to inhibiting the early replication of influenza virus, gGBP1 also inhibited the production of CCL2 and CXCL10 cytokines induced by the influenza virus as well as the proliferation of mononuclear macrophages induced by these cytokines. These findings further confirmed that gGBP1 inhibited the production of cytokines through its GTPase activity and cell proliferation through its C-terminal α-helix structure. This study revealed the effect of gGBP1 on the production of cellular inflammatory factors during influenza virus infection and determined the key amino acid residues that assist in the inhibitory processes mediated by gGBP1.

3.
Front Cell Infect Microbiol ; 14: 1341332, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38746783

RESUMO

Introduction: The Crimean-Congo hemorrhagic fever virus (CCHFV), the most geographically widespread tick-borne virus, is endemic in Africa, Eastern Europe and Asia, with infection resulting in mortality in up to 30% of cases. Currently, there are no approved vaccines or effective therapies available for CCHF. The CCHFV should only be manipulated in the BSL-4 laboratory, which has severely hampered basic seroprevalence studies. Methods: In the present study, two antibody detection methods in the forms of an enzyme-linked immunosorbent assay (ELISA) and a surrogate virus neutralization test (sPVNT) were developed using a recombinant glycoprotein (rGP) and a vesicular stomatitis virus (VSV)-based virus bearing the CCHFV recombinant glycoprotein (rVSV/CCHFV) in a biosafety level 2 (BSL-2) laboratory, respectively. Results: The rGP-based ELISA and rVSV/CCHFV-based sVNT were established by using the anti-CCHFV pre-GC mAb 11E7, known as a broadly cross-reactive, potently neutralizing antibody, and their applications as diagnostic antigens were validated for the specific detection of CCHFV IgG and neutralizing antibodies in experimental animals. In two tests, mAb clone 11E7 (diluted at 1:163840 or 512) still displayed positive binding and neutralization, and the presence of antibodies (IgG and neutralizing) against the rGP and rVSV/CCHFV was also determined in the sera from the experimental animals. Both mAb 11E7 and animal sera showed a high reactivity to both antigens, indicating that bacterially expressed rGP and rVSV/CCHFV have good immunoreactivity. Apart from establishing two serological testing methods, their results also demonstrated an imperfect correlation between IgG and neutralizing antibodies. Discussion: Within this limited number of samples, the rGP and rVSV/CCHFV could be safe and convenient tools with significant potential for research on specific antibodies and serological samples.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Ensaio de Imunoadsorção Enzimática , Vírus da Febre Hemorrágica da Crimeia-Congo , Febre Hemorrágica da Crimeia , Imunoglobulina G , Testes de Neutralização , Vírus da Febre Hemorrágica da Crimeia-Congo/imunologia , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Testes de Neutralização/métodos , Ensaio de Imunoadsorção Enzimática/métodos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Febre Hemorrágica da Crimeia/diagnóstico , Febre Hemorrágica da Crimeia/imunologia , Animais , Humanos , Glicoproteínas/imunologia , Testes Sorológicos/métodos , Proteínas Recombinantes/imunologia , Camundongos , Anticorpos Monoclonais/imunologia
4.
Viruses ; 16(4)2024 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-38675892

RESUMO

Canine distemper virus (CDV) can cause fatal infections in giant pandas. Vaccination is crucial to prevent CDV infection in giant pandas. In this study, two bacterium-like particle vaccines F3-GEM and H4-GEM displaying the trimeric F protein or tetrameric H protein of CDV were constructed based on the Gram-positive enhanced-matrix protein anchor (GEM-PA) surface display system. Electron microscopy and Western blot results revealed that the F or H protein was successfully anchored on the surface of GEM particles. Furthermore, one more bacterium-like particle vaccine F3 and H4-GEM was also designed, a mixture consisting of F3-GEM and H4-GEM at a ratio of 1:1. To evaluate the effect of the three vaccines, mice were immunized with F3-GEM, H4-GEM or F3 and H4-GEM. It was found that the level of IgG-specific antibodies and neutralizing antibodies in the F3 and H4-GEM group was higher than the other two groups. Additionally, F3 and H4-GEM also increased the secretion of Th1-related and Th2-related cytokines. Moreover, F3 and H4-GEM induce IgG and neutralizing antibodies' response in dogs. Conclusions: In summary, F3 and H4-GEM can provoke better immune responses to CDV in mice and dogs. The bacterium-like particle vaccine F3 and H4-GEM might be a potential vaccine candidate for giant pandas against CDV infection.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Vírus da Cinomose Canina , Cinomose , Vacinas Virais , Animais , Vírus da Cinomose Canina/imunologia , Cães , Camundongos , Cinomose/prevenção & controle , Cinomose/imunologia , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Feminino , Imunoglobulina G/sangue , Vacinas de Partículas Semelhantes a Vírus/imunologia , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/genética , Camundongos Endogâmicos BALB C , Citocinas/metabolismo , Vacinação
5.
Virus Res ; 345: 199378, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38643857

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a significant threat to human health globally. It is crucial to develop a vaccine to reduce the effect of the virus on public health, economy, and society and regulate the transmission of SARS-CoV-2. Influenza B virus (IBV) can be used as a vector that does not rely on the current circulating influenza A strains. In this study, we constructed an IBV-based vector vaccine by inserting a receptor-binding domain (RBD) into a non-structural protein 1 (NS1)-truncated gene (rIBV-NS110-RBD). Subsequently, we assessed its safety, immunogenicity, and protective efficacy against SARS-CoV-2 in mice, and observed that it was safe in a mouse model. Intranasal administration of a recombinant rIBV-NS110-RBD vaccine induced high levels of SARS-CoV-2-specific IgA and IgG antibodies and T cell-mediated immunity in mice. Administering two doses of the intranasal rIBV-NS110-RBD vaccine significantly reduced the viral load and lung damage in mice. This novel IBV-based vaccine offers a novel approach for controlling the SARS-CoV-2 pandemic.


Assuntos
Anticorpos Antivirais , Vacinas contra COVID-19 , COVID-19 , Vírus da Influenza B , Camundongos Endogâmicos BALB C , SARS-CoV-2 , Vacinas Atenuadas , Animais , Camundongos , Vírus da Influenza B/imunologia , Vírus da Influenza B/genética , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , SARS-CoV-2/imunologia , SARS-CoV-2/genética , COVID-19/prevenção & controle , COVID-19/imunologia , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas contra COVID-19/imunologia , Vacinas contra COVID-19/administração & dosagem , Feminino , Administração Intranasal , Humanos , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/genética , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/genética , Imunoglobulina A/sangue , Modelos Animais de Doenças , Imunoglobulina G/sangue , Carga Viral , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia
6.
Virol Sin ; 2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38556051

RESUMO

The Ebola virus (EBOV) is a member of the Orthoebolavirus genus, Filoviridae family, which causes severe hemorrhagic diseases in humans and non-human primates (NHPs), with a case fatality rate of up to 90%. The development of countermeasures against EBOV has been hindered by the lack of ideal animal models, as EBOV requires handling in biosafety level (BSL)-4 facilities. Therefore, accessible and convenient animal models are urgently needed to promote prophylactic and therapeutic approaches against EBOV. In this study, a recombinant vesicular stomatitis virus expressing Ebola virus glycoprotein (VSV-EBOV/GP) was constructed and applied as a surrogate virus, establishing a lethal infection in hamsters. Following infection with VSV-EBOV/GP, 3-week-old female Syrian hamsters exhibited disease signs such as weight loss, multi-organ failure, severe uveitis, high viral loads, and developed severe systemic diseases similar to those observed in human EBOV patients. All animals succumbed at 2-3 days post-infection (dpi). Histopathological changes indicated that VSV-EBOV/GP targeted liver cells, suggesting that the tissue tropism of VSV-EBOV/GP was comparable to wild-type EBOV (WT EBOV). Notably, the pathogenicity of the VSV-EBOV/GP was found to be species-specific, age-related, gender-associated, and challenge route-dependent. Subsequently, equine anti-EBOV immunoglobulins and a subunit vaccine were validated using this model. Overall, this surrogate model represents a safe, effective, and economical tool for rapid preclinical evaluation of medical countermeasures against EBOV under BSL-2 conditions, which would accelerate technological advances and breakthroughs in confronting Ebola virus disease.

7.
Antiviral Res ; 225: 105854, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38447647

RESUMO

The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants with frequent mutations has seriously damaged the effectiveness of the 2019 coronavirus disease (COVID-19) vaccine. There is an urgent need to develop a broad-spectrum vaccine while elucidating the underlying immune mechanisms. Here, we developed a SARS-CoV-2 virus-like particles (VLPs) vaccine based on the Canarypox-virus vector (ALVAC-VLPs) using CRISPR/Cas9. Immunization with ALVAC-VLPs showed the effectively induce SARS-CoV-2 specific T and B cell responses to resist the lethal challenge of mouse adaptive strains. Notably, ALVAC-VLPs conferred protection in golden hamsters against SARS-CoV-2 Wuhan-Hu-1 (wild-type, WT) and variants (Beta, Delta, Omicron BA.1, and BA.2), as evidenced by the prevention of weight loss, reduction in lung and turbinate tissue damage, and decreased viral load. Further investigation into the mechanism of immune response induced by ALVAC-VLPs revealed that toll-like receptor 4 (TLR4) mediates the recruitment of dendritic cells (DCs) to secondary lymphoid organs, thereby initiating follicle assisted T (Tfh) cell differentiation, the proliferation of germinal center (GC) B cells and plasma cell production. These findings demonstrate the immunogenicity and efficacy of the safe ALVAC-VLPs vaccine against SARS-CoV-2 and provide valuable insight into the development of COVID-19 vaccine strategies.


Assuntos
COVID-19 , Vacinas de Partículas Semelhantes a Vírus , Camundongos , Animais , Humanos , SARS-CoV-2 , Vacinas contra COVID-19 , Sistemas CRISPR-Cas , Edição de Genes , Anticorpos Antivirais , Anticorpos Neutralizantes
8.
Int J Biol Macromol ; 266(Pt 1): 131257, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38554908

RESUMO

The infected wounds pose one of the major threats to human health today. To address this issue, it is necessary to develop innovative wound dressings with superior antibacterial activity and other properties. Due to its potent antibacterial, antioxidant, and immune-boosting properties, epigallocatechin gallate (EGCG) has been widely utilized. In this study, a multifunctional curdlan hydrogel loading EGCG (Cur-EGCGH3) was designed. Cur-EGCGH3 exhibited excellent physicochemical properties, good biocompatibility, hemostatic, antibacterial, and antioxidant activities. Also, ELISA data showed that Cur-EGCGH3 stimulated macrophages to secrete pro-inflammatory and pro-regenerative cytokines. Cell scratch results indicated that Cur-EGCGH3 promoted the migration of NIH3T3 and HUVECs. In vivo experiments confirmed that Cur-EGCGH3 could inhibit bacterial infection of the infected wounds, accelerate hemostasis, and promote epithelial regeneration and collagen deposition. These results demonstrated that Cur-EGCGH3 holds promise for promoting healing of the infected wounds.


Assuntos
Antibacterianos , Catequina , Catequina/análogos & derivados , Hemostáticos , Hidrogéis , Cicatrização , beta-Glucanas , Catequina/farmacologia , Catequina/química , Animais , Cicatrização/efeitos dos fármacos , Camundongos , beta-Glucanas/química , beta-Glucanas/farmacologia , Hidrogéis/química , Hidrogéis/farmacologia , Antibacterianos/farmacologia , Antibacterianos/química , Humanos , Células NIH 3T3 , Hemostáticos/farmacologia , Hemostáticos/química , Infecção dos Ferimentos/tratamento farmacológico , Infecção dos Ferimentos/microbiologia , Antioxidantes/farmacologia , Antioxidantes/química , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos
9.
Int J Biol Macromol ; 264(Pt 2): 130820, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38484812

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and its variants has resulted in global economic losses and posed a threat to human health. The pandemic highlights the urgent need for an efficient, easily producible, and broad-spectrum vaccine. Here, we present a potentially universal strategy for the rapid and general design of vaccines, focusing on the design and testing of omicron BA.5 RBD-conjugated self-assembling ferritin nanoparticles (NPs). The covalent bonding of RBD-Fc to protein A-ferritin was easily accomplished through incubation, resulting in fully multivalent RBD-conjugated NPs that exhibited high structural uniformity, stability, and efficient assembly. The ferritin nanoparticle vaccine synergistically stimulated the innate immune response, Tfh-GCB-plasma cell-mediated activation of humoral immunity and IFN-γ-driven cellular immunity. This nanoparticle vaccine induced a high level of cross-neutralizing responses and protected golden hamsters challenged with multiple mutant strains from infection-induced clinical disease, providing a promising strategy for broad-spectrum vaccine development for SARS-CoV-2 prophylaxis. In conclusion, the nanoparticle conjugation platform holds promise for its potential universality and competitive immunization efficacy and is expected to facilitate the rapid manufacturing and broad application of next-generation vaccines.


Assuntos
COVID-19 , Nanopartículas , Animais , Cricetinae , Humanos , SARS-CoV-2 , COVID-19/prevenção & controle , Vacinas contra COVID-19 , Imunidade Inata , Ferritinas/genética , Nanovacinas , Anticorpos Neutralizantes , Anticorpos Antivirais
10.
J Med Microbiol ; 73(2)2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38420890

RESUMO

Introduction. Inappropriate use of antibiotics and inadequate therapeutic regimens for early-stage pulmonary infections are major contributors to increased prevalence of complications and mortality. Moreover, due to the limitations in sensitivity of conventional testing, there is an urgent need for more diagnostically efficient methods for the detection and characterization of pathogens in pulmonary infections.Hypothesis/Gap Statement. Metagenomic next-generation sequencing (mNGS) can contribute to the diagnosis and management of pulmonary infections.Aim. This study aimed to evaluate the clinical application and value of mNGS in the diagnosis of clinically suspected pulmonary infections by comparing with conventional testing.Methodology. In this study, the diagnosis performance of mNGS was evaluated using bronchoalveolar lavage fluid (BALF) samples from 143 patients with suspected lung infections. First, we conducted a prospective study on 31 patients admitted to Yuebei People's Hospital Affiliated to Shantou University Medical College to investigate the clinical value. Then a retrospective analysis was performed by including more patients (n=112) to reduce the random error. Pathogens were detected by mNGS and conventional methods (culture and PCR). Then, the types and cases of detected pathogens, as well as the specificity and sensitivity, were compared between the two methods. We evaluated the performance of mNGS in detecting bacterial, fungal, viral and mixed infections in BALF. The effect of disease severity in pulmonary infections on the integrity of mNGS pathogen detection was also explored.Results. The mNGS provided an earlier and more comprehensive pathogen profile than conventional testing, which in turn prompted a change in clinical medication, which led to improvement in eight patients (8/31=25.81 %) in the presence of other serious comorbidities. In a retrospective analysis, mNGS was much more sensitive than conventional testing in the diagnosis of pulmonary infections (95.33 % vs. 55.56 %; P<0.001), with a 39.77 % increase in sensitivity. The detection rate of mNGS for mixed infections was significantly higher than that of conventional testing methods for both common and severe pneumonia (48/67=71.64 % vs. 12/52=23.08 %, P<0.001; 44/59=74.58 % vs. 11/59=18.64 %, P<0.0001).Conclusion. The sensitivity of mNGS in the diagnosis of pathogenic microorganisms in pulmonary infections far exceeds that of conventional culture tests. As a complementary method to conventional methods, mNGS can help improve the diagnosis of pulmonary infections. In addition, mNGS pathogen integrity detection rate was similar in common and severe pneumonia. We recommend the prompt use of mNGS when mixed or rare pathogen infections are suspected, especially in immunocompromised individuals and/or critically ill individuals.


Assuntos
Bacteriófagos , Coinfecção , Pneumonia , Humanos , Líquido da Lavagem Broncoalveolar , Estudos Prospectivos , Estudos Retrospectivos , Sequenciamento de Nucleotídeos em Larga Escala , Metagenômica , Sensibilidade e Especificidade
11.
Vaccines (Basel) ; 11(12)2023 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-38140162

RESUMO

Nipah virus (NiV) causes severe, lethal encephalitis in humans and pigs. However, there is no licensed vaccine available to prevent NiV infection. In this study, we used the reverse genetic system based on the attenuated rabies virus strain SRV9 to construct two recombinant viruses, rSRV9-NiV-F and rSRV9-NiV-G, which displayed the NiV envelope glycoproteins F and G, respectively. Following three immunizations in BALB/c mice, the inactivated rSRV9-NiV-F and rSRV9-NiV-G alone or in combination, mixed with the adjuvants ISA 201 VG and poly (I:C), were able to induce the antigen-specific cellular and Th1-biased humoral immune responses. The specific antibodies against rSRV9-NiV-F and rSRV9-NiV-G had reactivity with two constructed bacterial-like particles displaying the F and G antigens of NiV. These data demonstrate that rSRV9-NiV-F or rSRV9-NiV-G has the potential to be developed into a promising vaccine candidate against NiV infection.

12.
J Virol ; 97(11): e0110123, 2023 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-37916835

RESUMO

IMPORTANCE: Clade 2.3.4.4 H5Nx avian influenza viruses (AIVs) have circulated globally and caused substantial economic loss. Increasing numbers of humans have been infected with Clade 2.3.4.4 H5N6 AIVs in recent years. Only a few human influenza vaccines have been licensed to date. However, the licensed live attenuated influenza virus vaccine exhibited the potential of being recombinant with the wild-type influenza A virus (IAV). Therefore, we developed a chimeric cold-adapted attenuated influenza vaccine based on the Clade 2.3.4.4 H5 AIVs. These H5 vaccines demonstrate the advantage of being non-recombinant with circulated IAVs in the future influenza vaccine study. The findings of our current study reveal that these H5 vaccines can induce cross-reactive protective efficacy in mice and ferrets. Our H5 vaccines may provide a novel option for developing human-infected Clade 2.3.4.4 H5 AIV vaccines.


Assuntos
Proteção Cruzada , Vírus da Influenza A , Vacinas contra Influenza , Infecções por Orthomyxoviridae , Animais , Camundongos , Anticorpos Antivirais , Furões , Influenza Aviária , Vacinas contra Influenza/genética , Vacinas Atenuadas , Infecções por Orthomyxoviridae/prevenção & controle
13.
Antiviral Res ; 220: 105765, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-38036065

RESUMO

Coronavirus disease 2019 (COVID-19) seriously threatens public health safety and the global economy, which warrant effective prophylactic and therapeutic approaches. Currently, vaccination and establishment of immunity have significantly reduced the severity and mortality of COVID-19. However, in regard to COVID-19 vaccines, the broad-spectrum protective efficacy against SARS-CoV-2 variants and the blocking of virus transmission need to be further improved. In this study, an optimum oral COVID-19 vaccine candidate, rVSVΔG-Sdelta, was selected from a panel of vesicular stomatitis virus (VSV)-based constructs bearing spike proteins from different SARS-CoV-2 strains. After chitosan modification, rVSVΔG-Sdelta induced both local and peripheral antibody response, particularly, broad-spectrum and long-lasting neutralizing antibodies against SARS-CoV-2 persisted for 1 year. Cross-protection against SARS-CoV-2 WT, Beta, Delta, BA.1, and BA.2 strains was achieved in golden hamsters, which presented as significantly reduced viral replication in the respiratory tract and alleviated pulmonary pathology post SARS-CoV-2 challenge. Overall, this study provides a convenient, oral-delivered, and effective oral mucosal vaccine against COVID-19, which would supplement pools and facilitate the distribution of COVID-19 vaccines.


Assuntos
COVID-19 , Quitosana , Animais , Cricetinae , Humanos , SARS-CoV-2 , Vacinas contra COVID-19 , Mesocricetus , COVID-19/prevenção & controle , Adjuvantes Imunológicos , Anticorpos Neutralizantes , Anticorpos Antivirais , Glicoproteína da Espícula de Coronavírus/genética
14.
FEBS Open Bio ; 13(10): 1831-1843, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37544014

RESUMO

Influenza virus is prone to seasonal spread and widespread outbreaks, which pose important challenges to public health security. Therefore, it is important to effectively prevent and treat influenza virus infection. Schisandra polysaccharide (SPJ) is a polysaccharide derived from the fruit of Schisandra chinensis (Turcz.) Baill. In this study, we evaluated the antiviral activity of SPJ in vitro and in vivo, especially against influenza A virus (IAV) infection. By analyzing SPJ structure and monosaccharide composition, the molecular weight of SPJ was determined to be 115.5 KD, and it is composed of galacturonic acid (89.4%), rhamnose (0.8%), galactose (4.4%), arabinose (3.8%), and glucose (1.7%). Immunofluorescence analysis showed that SPJ treatment reduced the positive rate of viral nucleoproteins in cells, indicating that the compound had an inhibitory effect on influenza virus replication. Furthermore, SPJ therapy improved the survival of infected mice. Lung virus titer assays indicated that SPJ treatment significantly reduced viral loading in the lung tissue of infected mice and alleviated the pathological damage caused by influenza virus infection. Moreover, SPJ reduced cytokine expression during influenza virus challenge. In conclusion, SPJ has anti-influenza virus effects and may have potential as an anti-influenza drug candidate in further clinical studies.

15.
Small ; 19(45): e2303542, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37431212

RESUMO

Rabies is a zoonotic neurological disease caused by the rabies virus (RABV) that is fatal to humans and animals. While several post-infection treatment have been suggested, developing more efficient and innovative antiviral methods are necessary due to the limitations of current therapeutic approaches. To address this challenge, a strategy combining photodynamic therapy and immunotherapy, using a photosensitizer (TPA-Py-PhMe) with high type I and type II reactive oxygen species (ROS) generation ability is proposed. This approach can inactivate the RABV by killing the virus directly and activating the immune response. At the cellular level, TPA-Py-PhMe can reduce the virus titer under preinfection prophylaxis and postinfection treatment, with its antiviral effect mainly dependent on ROS and pro-inflammatory factors. Intriguingly, when mice are injected with TPA-Py-PhMe and exposed to white light irradiation at three days post-infection, the onset of disease is delayed, and survival rates improved to some extent. Overall, this study shows that photodynamic therapy and immunotherapy open new avenues for future antiviral research.


Assuntos
Fotoquimioterapia , Vírus da Raiva , Raiva , Humanos , Animais , Camundongos , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Espécies Reativas de Oxigênio , Raiva/prevenção & controle , Raiva/tratamento farmacológico , Antivirais
16.
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
17.
Virology ; 585: 145-154, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37348143

RESUMO

The SARS-CoV-2 pandemic has continued for about three years since emerging in late December 2019, resulting in millions of deaths. Therefore, there is an urgent need to develop a safe and effective vaccine to control SARS-CoV-2. In this study, we developed a bacterium-like particle vaccine that displays the SARS-CoV-2 receptor binding domain (RBD) (named Trim-RBD-GEM) using the GEM-PA system. We evaluated the immunogenicity and protective efficacy of the Trim-RBD-GEM vaccine with the oil-in-water adjuvant AddaVax in C57BL/6 N mice intramuscularly. We found that Trim-RBD-GEM&AddaVax induced high levels of humoral immunity in C57BL/6 N mice. Additionally, the lung virus loads in the immunized group were significantly decreased compared to the adjuvant control and mock groups. Therefore, this vaccine provides protection against lethal infection in a C57BL/6 N mouse model. Our Trim-RBD-GEM&AddaVax vaccine is potentially a promising, rapid, and safe subunit vaccine for preventing and controlling SARS-CoV-2.


Assuntos
COVID-19 , Vacinas , Animais , Camundongos , Camundongos Endogâmicos C57BL , COVID-19/prevenção & controle , SARS-CoV-2/genética , Adjuvantes Imunológicos , Glicoproteína da Espícula de Coronavírus , Anticorpos Antivirais , Anticorpos Neutralizantes
18.
Signal Transduct Target Ther ; 8(1): 149, 2023 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-37029123

RESUMO

Human diseases, particularly infectious diseases and cancers, pose unprecedented challenges to public health security and the global economy. The development and distribution of novel prophylactic and therapeutic vaccines are the prioritized countermeasures of human disease. Among all vaccine platforms, viral vector vaccines offer distinguished advantages and represent prominent choices for pathogens that have hampered control efforts based on conventional vaccine approaches. Currently, viral vector vaccines remain one of the best strategies for induction of robust humoral and cellular immunity against human diseases. Numerous viruses of different families and origins, including vesicular stomatitis virus, rabies virus, parainfluenza virus, measles virus, Newcastle disease virus, influenza virus, adenovirus and poxvirus, are deemed to be prominent viral vectors that differ in structural characteristics, design strategy, antigen presentation capability, immunogenicity and protective efficacy. This review summarized the overall profile of the design strategies, progress in advance and steps taken to address barriers to the deployment of these viral vector vaccines, simultaneously highlighting their potential for mucosal delivery, therapeutic application in cancer as well as other key aspects concerning the rational application of these viral vector vaccines. Appropriate and accurate technological advances in viral vector vaccines would consolidate their position as a leading approach to accelerate breakthroughs in novel vaccines and facilitate a rapid response to public health emergencies.


Assuntos
Doenças Transmissíveis , Orthomyxoviridae , Vacinas Virais , Animais , Humanos , Vacinas Virais/genética , Vacinas Virais/uso terapêutico , Vetores Genéticos , Orthomyxoviridae/genética , Adenoviridae/genética
19.
Microbiol Spectr ; 11(3): e0307922, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37014208

RESUMO

Bax-interacting factor-1 (Bif-1) is a multifunctional protein involved in apoptosis, autophagy, and mitochondrial morphology. However, the associations between Bif-1 and viruses are poorly understood. As discrete Bif-1 isoforms are selectively expressed and exert corresponding effects, we evaluated the effects of neuron-specific/ubiquitous Bif-1 isoforms on rabies virus (RABV) proliferation. First, infection with the RABV CVS-11 strain significantly altered Bif-1 expression in mouse neuroblastoma (N2a) cells, and Bif-1 knockdown in turn promoted RABV replication. Overexpression of neuron-specific Bif-1 isoforms (Bif-1b/c/e) suppressed RABV replication. Moreover, our study showed that Bif-1c colocalized with LC3 and partially alleviated the incomplete autophagic flux induced by RABV. Taken together, our data reveal that neuron-specific Bif-1 isoforms impair the RABV replication process by abolishing autophagosome accumulation and blocking autophagic flux induced by the RABV CVS-11 strain in N2a cells. IMPORTANCE Autophagy can be triggered by viral infection and replication. Autophagosomes are generated and affect RABV replication, which differs by viral strain and infected cell type. Bax-interacting factor-1 (Bif-1) mainly has a proapoptotic function but is also involved in autophagosome formation. However, the association between Bif-1-involved autophagy and RABV infection remains unclear. In this study, our data reveal that a neuron-specific Bif-1 isoform, Bif-1c, impaired viral replication by unchoking autophagosome accumulation induced by RABV in N2a cells to a certain extent. Our study reveals for the first time that Bif-1 is involved in modulating autophagic flux and plays a crucial role in RABV replication, establishing Bif-1 as a potential therapeutic target for rabies.


Assuntos
Vírus da Raiva , Raiva , Animais , Camundongos , Vírus da Raiva/metabolismo , Proteína X Associada a bcl-2/metabolismo , Proteína X Associada a bcl-2/farmacologia , Autofagia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacologia , Proliferação de Células
20.
J Virol ; 97(3): e0001123, 2023 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-36877072

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the viral pathogen responsible for the worldwide coronavirus disease 2019 (COVID-19) pandemic. The novel SARS-CoV-2 ORF8 protein is not highly homologous with known proteins, including accessory proteins of other coronaviruses. ORF8 contains a 15-amino-acid signal peptide in the N terminus that localizes the mature protein to the endoplasmic reticulum. Oligomannose-type glycosylation has been identified at the N78 site. Here, the unbiased molecular functions of ORF8 are also demonstrated. Via an immunoglobulin-like fold in a glycan-independent manner, both exogenous and endogenous ORF8 interacts with human calnexin and HSPA5. The key ORF8-binding sites of Calnexin and HSPA5 are indicated on the globular domain and the core substrate-binding domain, respectively. ORF8 induces species-dependent endoplasmic reticulum stress-like responses in human cells exclusively via the IRE1 branch, including intensive HSPA5 and PDIA4 upregulation, with increases in other stress-responding effectors, including CHOP, EDEM and DERL3. ORF8 overexpression facilitates SARS-CoV-2 replication. Both stress-like responses and viral replication induced by ORF8 have been shown to result from triggering the Calnexin switch. Thus, ORF8 serves as a key unique virulence gene of SARS-CoV-2, potentially contributing to COVID-19-specific and/or human-specific pathogenesis. IMPORTANCE Although SARS-CoV-2 is basically regarded as a homolog of SARS-CoV, with their genomic structure and the majority of their genes being highly homologous, the ORF8 genes of SARS-CoV and SARS-CoV-2 are distinct. The SARS-CoV-2 ORF8 protein also shows little homology with other viral or host proteins and is thus regarded as a novel special virulence gene of SARS-CoV-2. The molecular function of ORF8 has not been clearly known until now. Our results reveal the unbiased molecular characteristics of the SARS-CoV-2 ORF8 protein and demonstrate that it induces rapidly generated but highly controllable endoplasmic reticulum stress-like responses and facilitates virus replication by triggering Calnexin in human but not mouse cells, providing an explanation for the superficially known in vivo virulence discrepancy of ORF8 between SARS-CoV-2-infected patients and mouse.


Assuntos
COVID-19 , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave , Humanos , Calnexina/genética , SARS-CoV-2/genética , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...