Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Virol ; 98(4): e0156523, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38445884

RESUMEN

The COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has posed a worldwide threat in the past 3 years. Although it has been widely and intensively investigated, the mechanism underlying the coronavirus-host interaction requires further elucidation, which may contribute to the development of new antiviral strategies. Here, we demonstrated that the host cAMP-responsive element-binding protein (CREB1) interacts with the non-structural protein 13 (nsp13) of SARS-CoV-2, a conserved helicase for coronavirus replication, both in cells and in lung tissues subjected to SARS-CoV-2 infection. The ATPase and helicase activity of viral nsp13 were shown to be potentiated by CREB1 association, as well as by Protein kinase A (PKA)-mediated CREB1 activation. SARS-CoV-2 replication is significantly suppressed by PKA Cα, cAMP-activated protein kinase catalytic subunit alpha (PRKACA), and CREB1 knockdown or inhibition. Consistently, the CREB1 inhibitor 666-15 has shown significant antiviral effects against both the WIV04 strain and the Omicron strain of the SARS-CoV-2. Our findings indicate that the PKA-CREB1 signaling axis may serve as a novel therapeutic target against coronavirus infection. IMPORTANCE: In this study, we provide solid evidence that host transcription factor cAMP-responsive element-binding protein (CREB1) interacts directly with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) helicase non-structural protein 13 (nsp13) and potentiate its ATPase and helicase activity. And by live SARS-CoV-2 virus infection, the inhibition of CREB1 dramatically impairs SARS-CoV-2 replication in vivo. Notably, the IC50 of CREB1 inhibitor 666-15 is comparable to that of remdesivir. These results may extend to all highly pathogenic coronaviruses due to the conserved nsp13 sequences in the virus.


Asunto(s)
ARN Polimerasa Dependiente de ARN de Coronavirus , Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico , Interacciones Microbiota-Huesped , SARS-CoV-2 , Proteínas no Estructurales Virales , Replicación Viral , Humanos , Adenosina Trifosfatasas/metabolismo , Antivirales/farmacología , ARN Polimerasa Dependiente de ARN de Coronavirus/metabolismo , COVID-19/virología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/antagonistas & inhibidores , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/deficiencia , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , ADN Helicasas/metabolismo , Concentración 50 Inhibidora , ARN Helicasas/metabolismo , SARS-CoV-2/clasificación , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/enzimología , SARS-CoV-2/crecimiento & desarrollo , Transducción de Señal/efectos de los fármacos , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/efectos de los fármacos , Femenino , Animales , Ratones
2.
Mol Ther ; 32(4): 1033-1047, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38341613

RESUMEN

As the world continues to confront severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), respiratory syncytial virus (RSV) is also causing severe respiratory illness in millions of infants, elderly individuals, and immunocompromised people globally. Exacerbating the situation is the fact that co-infection with multiple viruses is occurring, something which has greatly increased the clinical severity of the infections. Thus, our team developed a bivalent vaccine that delivered mRNAs encoding SARS-CoV-2 Omicron spike (S) and RSV fusion (F) proteins simultaneously, SF-LNP, which induced S and F protein-specific binding antibodies and cellular immune responses in BALB/c mice. Moreover, SF-LNP immunization effectively protected BALB/c mice from RSV infection and hamsters from SARS-CoV-2 Omicron infection. Notably, our study pointed out the antigenic competition problem of bivalent vaccines and provided a solution. Overall, our results demonstrated the potential of preventing two infectious diseases with a single vaccine and provided a paradigm for the subsequent design of multivalent vaccines.


Asunto(s)
COVID-19 , Infecciones por Virus Sincitial Respiratorio , Vacunas contra Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , Humanos , Ratones , Lactante , Cricetinae , Animales , Anciano , Vacunas de ARNm , Vacunas Combinadas , Anticuerpos Antivirales , Vacunas contra Virus Sincitial Respiratorio/genética , Proteínas Virales de Fusión/genética , COVID-19/prevención & control , SARS-CoV-2/genética , Virus Sincitial Respiratorio Humano/genética , Infecciones por Virus Sincitial Respiratorio/prevención & control , Anticuerpos Neutralizantes
3.
Mol Ther ; 32(6): 1779-1789, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38659224

RESUMEN

Since the outbreak of monkeypox (mpox) in 2022, widespread concern has been placed on imposing an urgent demand for specific vaccines that offer safer and more effective protection. Using an efficient and scalable circular RNA (circRNA) platform, we constructed four circRNA vaccines that could induce robust neutralizing antibodies as well as T cell responses by expressing different surface proteins of mpox virus (MPXV), resulting in potent protection against vaccinia virus (VACV) in mice. Strikingly, the combination of the four circular RNA vaccines demonstrated the best protection against VACV challenge among all the tested vaccines. Our study provides a favorable approach for developing MPXV-specific vaccines by using a circular mRNA platform and opens up novel avenues for future vaccine research.


Asunto(s)
Anticuerpos Neutralizantes , Monkeypox virus , ARN Circular , Virus Vaccinia , Animales , Ratones , Virus Vaccinia/genética , Virus Vaccinia/inmunología , ARN Circular/genética , Anticuerpos Neutralizantes/inmunología , Monkeypox virus/inmunología , Monkeypox virus/genética , Anticuerpos Antivirales/inmunología , Vaccinia/prevención & control , Vaccinia/inmunología , Mpox/prevención & control , Mpox/inmunología , Vacunas Virales/inmunología , Vacunas Virales/genética , Humanos , Modelos Animales de Enfermedad , Femenino , Linfocitos T/inmunología , Linfocitos T/metabolismo
4.
J Virol ; 96(12): e0041222, 2022 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-35652658

RESUMEN

SARS-CoV-2 is the causative agent of the ongoing pandemic of coronavirus disease 2019 (COVID-19) and poses a significant threat to global health. N protein (NP), which is a major pathogenic protein among betacoronaviruses, binds to the viral RNA genome to allow viral genome packaging and viral particle release. Recent studies showed that NP antagonizes interferon (IFN) induction and mediates phase separation. Using live SARS-CoV-2 viruses, this study provides solid evidence showing that SARS-CoV-2 NP associates with G3BP1 and G3BP2 in vitro and in vivo. NPSARS-CoV-2 could efficiently suppress G3BP-mediated SG formation and potentiate viral infection by overcoming G3BP1-mediated antiviral innate immunity. G3BP1 conditional knockout mice (g3bp1fl/fL, Sftpc-Cre) exhibit significantly higher lung viral loads after SARS-CoV-2 infection than wild-type mice. Our findings contribute to the growing body of knowledge regarding the pathogenicity of NPSARS-CoV-2 and provide insight into new therapeutics targeting NPSARS-CoV-2. IMPORTANCE In this study, by in vitro assay and live SARS-CoV-2 virus infection, we provide solid evidence that the SARS-CoV-2 NP associates with G3BP1 and G3BP2 in vitro and in vivo. NPSARS-CoV-2 could efficiently suppress G3BP-mediated SG formation and potentiate viral infection by overcoming antiviral innate immunity mediated by G3BP1 in A549 cell lines and G3BP1 conditional knockout mice (g3bp1-cKO) mice, which provide in-depth evidence showing the mechanism underlying NP-related SARS-CoV-2 pathogenesis through G3BPs.


Asunto(s)
COVID-19 , Proteínas de la Nucleocápside de Coronavirus , Proteínas de Unión a Poli-ADP-Ribosa , SARS-CoV-2 , Replicación Viral , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , COVID-19/inmunología , COVID-19/virología , Proteínas de la Nucleocápside de Coronavirus/metabolismo , ADN Helicasas/metabolismo , Interacciones Microbiota-Huesped/inmunología , Ratones , Fosfoproteínas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , ARN Helicasas/metabolismo , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Gránulos de Estrés , Replicación Viral/genética
5.
Virol J ; 20(1): 75, 2023 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-37081549

RESUMEN

BACKGROUND: The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes non-symptomatic infection, mild influenza-like symptoms to pneumonia, severe acute respiratory distress syndrome, and even death, reflecting different clinical symptoms of viral infection. However, the mechanism of its pathogenicity remains unclear. Host-specific traits have a breakthrough significance for studying the pathogenicity of SARS-CoV-2. We previously reported SARS-CoV-2/BMA8, a mouse-adapted strain, was lethal to aged BALB/c mice but not to aged C57BL/6N mice. Here, we further investigate the differences in pathogenicity of BMA8 strain against wild-type aged C57BL/6N and BALB/c mice. METHODS: Whole blood and tissues were collected from mice before and after BMA8 strain infection. Viral replication and infectivity were assessed by detection of viral RNA copies and viral titers; the degree of inflammation in mice was tested by whole blood cell count, ELISA and RT-qPCR assays; the pathogenicity of SARS-CoV-2/BMA8 in mice was measured by Histopathology and Immunohistochemistry; and the immune level of mice was evaluated by flow cytometry to detect the number of CD8+ T cells. RESULTS: Our results suggest that SARS-CoV-2/BMA8 strain caused lower pathogenicity and inflammation level in C57BL/6N mice than in BALB/c mice. Interestingly, BALB/c mice whose MHC class I haplotype is H-2Kd showed more severe pathogenicity after infection with BMA8 strain, while blockade of H-2Kb in C57BL/6N mice was also able to cause this phenomenon. Furthermore, H-2Kb inhibition increased the expression of cytokines/chemokines and accelerated the decrease of CD8+ T cells caused by SARS-CoV-2/BMA8 infection. CONCLUSIONS: Taken together, our work shows that host MHC molecules play a crucial role in the pathogenicity differences of SARS-CoV-2/BMA8 infection. This provides a more profound insight into the pathogenesis of SARS-CoV-2, and contributes enlightenment and guidance for controlling the virus spread.


Asunto(s)
COVID-19 , SARS-CoV-2 , Ratones , Animales , Linfocitos T CD8-positivos , Virulencia , COVID-19/patología , Ratones Endogámicos C57BL , Ratones Endogámicos BALB C , Inflamación , Pulmón/patología , Modelos Animales de Enfermedad
6.
BMC Vet Res ; 17(1): 172, 2021 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-33892731

RESUMEN

BACKGROUND: Canine distemper virus (CDV) is an enveloped negative-strand RNA virus that exhibits a high mutation rate and continuously expands the range of hosts. Notably, CDV has infected giant panda with spill over from viral reservoirs in canines. Giant pandas (Ailuropoda melanoleuca), especially captive pandas, are known to be susceptible to natural infection with CDV. The high fatality rate of CDV poses a serious threat to the safety of the giant panda population. However, vaccines or drugs for canine distemper in giant pandas have not been developed to date. Therefore, a rapid test that can achieve accurate onsite detection of CDV is important to enable the timely implementation of control measures. In this study, we established a nucleic acid visualization assay for targeting the CDV N gene by using combines reverse transcription recombinase polymerase amplification with a closed vertical flow visualization strip (RT-RPA-VF). RESULTS: The RT-RPA-VF assay does not require sophisticated equipment, and it was determined to provide rapid detection at 35 °C for 30 min, while the limit of detection was 5 × 101 copies/µl RNA transcripts and 100.5 TCID50 ml- 1 viruses. The results showed that the assay was high specific to CDV and had no cross-reactivity with other viruses infecting the giant panda. Compared with RT-qPCR, RT-RPA-VF assay had a sensitivity of 100% and a specificity of 100% in 29 clinical samples. The coincidence rate between RT-RPA-VF and RT-qPCR was 100% (kappa = 1), indicating that the RT-RPA-VF assay possessed good diagnostic performance on clinical samples. CONCLUSIONS: The RT-RPA-VF provides a novel alternative for the simple, sensitive, and specific identification of CDV and showed great potential for point of care diagnostics for captive and wild giant panda.


Asunto(s)
Virus del Moquillo Canino/genética , Virus del Moquillo Canino/aislamiento & purificación , Moquillo/diagnóstico , Técnicas de Amplificación de Ácido Nucleico/veterinaria , Ursidae/virología , Animales , Moquillo/virología , Técnicas de Amplificación de Ácido Nucleico/métodos , ARN Viral , Reacción en Cadena en Tiempo Real de la Polimerasa/veterinaria , Transcripción Reversa , Sensibilidad y Especificidad
7.
Nat Commun ; 15(1): 2987, 2024 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-38582870

RESUMEN

Nipah virus (NiV) is a World Health Organization priority pathogen and there are currently no approved drugs for clinical immunotherapy. Through the use of a naïve human phage-displayed Fab library, two neutralizing antibodies (NiV41 and NiV42) targeting the NiV receptor binding protein (RBP) were identified. Following affinity maturation, antibodies derived from NiV41 display cross-reactivity against both NiV and Hendra virus (HeV), whereas the antibody based on NiV42 is only specific to NiV. Results of immunogenetic analysis reveal a correlation between the maturation of antibodies and their antiviral activity. In vivo testing of NiV41 and its mature form (41-6) show protective efficacy against a lethal NiV challenge in hamsters. Furthermore, a 2.88 Å Cryo-EM structure of the tetrameric RBP and antibody complex demonstrates that 41-6 blocks the receptor binding interface. These findings can be beneficial for the development of antiviral drugs and the design of vaccines with broad spectrum against henipaviruses.


Asunto(s)
Infecciones por Henipavirus , Virus Nipah , Humanos , Anticuerpos Neutralizantes/metabolismo , Virus Nipah/metabolismo , Anticuerpos Antivirales
8.
J Control Release ; 366: 479-493, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38184234

RESUMEN

mRNA-based vaccines and therapeutic agents hold great promise in prevention and treatment of human diseases, yet high percentage of systemic adverse effect in clinic remains a big safety concern. One major potential cause is a high level of leakage of the locally inoculated mRNA vaccine nanoparticles into circulation. We have screened and optimized a core-shell structured lipopolyplex (LPP) formulation for mRNA with a tissue-retention property. Upon intramuscular inoculation, the mRNA-encapsulated LPP nanoparticles were preferentially taken up by the phagocytic antigen-presentation cells, and potently promoted dendritic cell maturation. We applied the new formulation to prepare a prophylactic vaccine for SARS-CoV-2, and observed potent humoral and cellular immune responses from the vaccine in both murine models and non-human primates. More importantly, the vaccine demonstrated a benign safety profile in non-human primates, with limited side effects after repeated treatment with high dosages of LPP/mRNA. Taken together, the inoculation site-retained vaccine formulation serves as a promising vehicle for mRNA vaccines and therapeutic agents.


Asunto(s)
COVID-19 , Vacunas de ARNm , Humanos , Animales , Ratones , SARS-CoV-2/genética , Vacunas contra la COVID-19 , COVID-19/prevención & control , Presentación de Antígeno , ARN Mensajero , Primates , Anticuerpos Antivirales , Anticuerpos Neutralizantes
9.
Virus Res ; 345: 199378, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38643857

RESUMEN

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.


Asunto(s)
Anticuerpos Antivirales , Vacunas contra la COVID-19 , COVID-19 , Virus de la Influenza B , Ratones Endogámicos BALB C , SARS-CoV-2 , Vacunas Atenuadas , Animales , Ratones , Virus de la Influenza B/inmunología , Virus de la Influenza B/genética , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , SARS-CoV-2/inmunología , SARS-CoV-2/genética , COVID-19/prevención & control , COVID-19/inmunología , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/administración & dosificación , Femenino , Administración Intranasal , Humanos , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Inmunoglobulina A/sangre , Modelos Animales de Enfermedad , Inmunoglobulina G/sangre , Carga Viral , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología
10.
Cell Rep ; 43(6): 114269, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38787725

RESUMEN

The 2022 mpox outbreak led the World Health Organization (WHO) to declare it a public health emergency of international concern (PHEIC). There is a need to develop more effective and safer mpox virus (MPXV)-specific vaccines in response to the mpox epidemic. The mRNA vaccine is a promising platform to protect against MPXV infection. In this study, we construct two bivalent MPXV mRNA vaccines, designated LBA (B6R-A29L) and LAM (A35R-M1R), and a quadrivalent mRNA vaccine, LBAAM (B6R-A35R-A29L-M1R). The immunogenicity and protective efficacy of these vaccines alone or in combination were evaluated in a lethal mouse model. All mRNA vaccine candidates could elicit potential antigen-specific humoral and cellular immune responses and provide protection against vaccinia virus (VACV) infection. The protective effect of the combination of two bivalent mRNA vaccines and the quadrivalent vaccine was superior to that of the individual bivalent mRNA vaccine. Our study provides valuable insights for the development of more efficient and safer mRNA vaccines against mpox.


Asunto(s)
Virus Vaccinia , Vacunas de ARNm , Animales , Virus Vaccinia/inmunología , Virus Vaccinia/genética , Ratones , Femenino , Vacunas de ARNm/inmunología , Humanos , Ratones Endogámicos BALB C , Mpox/prevención & control , Mpox/inmunología , Vaccinia/inmunología , Vaccinia/prevención & control , Anticuerpos Antivirales/inmunología , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Mensajero/inmunología , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Inmunidad Humoral
11.
Nat Commun ; 15(1): 4330, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38773072

RESUMEN

The Hendra and Nipah viruses (HNVs) are highly pathogenic pathogens without approved interventions for human use. In addition, the interaction pattern between the attachment (G) and fusion (F) glycoproteins required for virus entry remains unclear. Here, we isolate a panel of Macaca-derived G-specific antibodies that cross-neutralize HNVs via multiple mechanisms. The most potent antibody, 1E5, confers adequate protection against the Nipah virus challenge in female hamsters. Crystallography demonstrates that 1E5 has a highly similar binding pattern to the receptor. In cryo-electron microscopy studies, the tendency of 1E5 to bind to the upper or lower heads results in two distinct quaternary structures of G. Furthermore, we identify the extended outer loop ß1S2-ß1S3 of G and two pockets on the apical region of fusion (F) glycoprotein as the essential sites for G-F interactions. This work highlights promising drug candidates against HNVs and contributes deeper insights into the viruses.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Microscopía por Crioelectrón , Infecciones por Henipavirus , Proteínas Virales de Fusión , Animales , Anticuerpos Neutralizantes/inmunología , Femenino , Anticuerpos Antivirales/inmunología , Infecciones por Henipavirus/virología , Infecciones por Henipavirus/inmunología , Proteínas Virales de Fusión/inmunología , Proteínas Virales de Fusión/química , Humanos , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/química , Virus Nipah/inmunología , Internalización del Virus/efectos de los fármacos , Henipavirus/inmunología , Cricetinae , Reacciones Cruzadas/inmunología , Virus Hendra/inmunología , Macaca , Mesocricetus , Cristalografía por Rayos X
12.
Sci China Life Sci ; 67(7): 1502-1513, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38478297

RESUMEN

Various SARS-CoV-2-related coronaviruses have been increasingly identified in pangolins, showing a potential threat to humans. Here we report the infectivity and pathogenicity of the SARS-CoV-2-related virus, PCoV-GX/P2V, which was isolated from a Malayan pangolin (Manis javanica). PCoV-GX/P2V could grow in human hepatoma, colorectal adenocarcinoma cells, and human primary nasal epithelial cells. It replicated more efficiently in cells expressing human angiotensin-converting enzyme 2 (hACE2) as SARS-CoV-2 did. After intranasal inoculation to the hACE2-transgenic mice, PCoV-GX/P2V not only replicated in nasal turbinate and lungs, but also caused interstitial pneumonia, characterized by infiltration of mixed inflammatory cells and multifocal alveolar hemorrhage. Existing population immunity established by SARS-CoV-2 infection and vaccination may not protect people from PCoV-GX/P2V infection. These findings further verify the hACE2 utility of PCoV-GX/P2V by in vivo experiments using authentic viruses and highlight the importance for intensive surveillance to prevent possible cross-species transmission.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , COVID-19 , Ratones Transgénicos , Pangolines , SARS-CoV-2 , Animales , Humanos , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/genética , SARS-CoV-2/patogenicidad , SARS-CoV-2/genética , COVID-19/virología , Pangolines/virología , Ratones , Replicación Viral , Pulmón/virología , Pulmón/patología , Chlorocebus aethiops , Células Vero
13.
Front Microbiol ; 14: 1126533, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36846792

RESUMEN

Introduction: Rabies is a worldwide epidemic that poses a serious threat to global public health. At present, rabies in domestic dogs, cats, and some pets can be effectively prevented and controlled by intramuscular injection of rabies vaccine. But for some inaccessible animals, especially stray dogs, and wild animals, it is difficult to prevent with intramuscular injection. Therefore, it is necessary to develop a safe and effective oral rabies vaccine. Methods: We constructed recombinant Bacillus subtilis (B. subtilis) expressing two different strains of rabies virus G protein, named CotG-E-G and CotG-C-G, immunogenicity was studied in mice. Results: The results showed that CotG-E-G and CotG-C-G could significantly increase the specific SIgA titers in feces, serum IgG titers, and neutralizing antibodies. ELISpot experiments showed that CotG-E-G and CotG-C-G could also induce Th1 and Th2 to mediate the secretion of immune-related IFN-γ and IL-4. Collectively, our results suggested that recombinant B. subtilis CotG-E-G and CotG-C-G have excellent immunogenicity and are expected to be novel oral vaccine candidates for the prevention and control of wild animal rabies.

14.
J Pharm Anal ; 13(1): 11-23, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36313960

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced cytokine storms constitute the primary cause of coronavirus disease 19 (COVID-19) progression, severity, criticality, and death. Glucocorticoid and anti-cytokine therapies are frequently administered to treat COVID-19, but have limited clinical efficacy in severe and critical cases. Nevertheless, the weaknesses of these treatment modalities have prompted the development of anti-inflammatory therapy against this infection. We found that the broad-spectrum anti-inflammatory agent inosine downregulated proinflammatory interleukin (IL)-6, upregulated anti-inflammatory IL-10, and ameliorated acute inflammatory lung injury caused by multiple infectious agents. Inosine significantly improved survival in mice infected with SARS-CoV-2. It indirectly impeded TANK-binding kinase 1 (TBK1) phosphorylation by binding stimulator of interferon genes (STING) and glycogen synthase kinase-3ß (GSK3ß), inhibited the activation and nuclear translocation of the downstream transcription factors interferon regulatory factor (IRF3) and nuclear factor kappa B (NF-κB), and downregulated IL-6 in the sera and lung tissues of mice infected with lipopolysaccharide (LPS), H1N1, or SARS-CoV-2. Thus, inosine administration is feasible for clinical anti-inflammatory therapy against severe and critical COVID-19. Moreover, targeting TBK1 is a promising strategy for inhibiting cytokine storms and mitigating acute inflammatory lung injury induced by SARS-CoV-2 and other infectious agents.

15.
Virol Sin ; 38(6): 922-930, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37839549

RESUMEN

As one of the deadliest viruses, Ebola virus (EBOV) causes lethal hemorrhagic fevers in humans and nonhuman primates. The suppression of innate immunity leads to robust systemic virus replication of EBOV, leading to enhanced transmission. However, the mechanism of EBOV-host interaction is not fully understood. Here, we identified multiple dysregulated genes in early stage of EBOV infection through transcriptomic analysis, which are highly clustered to Jak-STAT signaling. EBOV VP35 and VP30 were found to inhibit type I interferon (IFN) signaling. Moreover, exogenous expression of VP35 blocks the phosphorylation of endogenous STAT1, and suppresses nuclear translocation of STAT1. Using serial truncated mutations of VP35, N-terminal 1-220 amino acid residues of VP35 were identified to be essential for blocking on type I IFN signaling. Remarkably, VP35 of EBOV suppresses type I IFN signaling more efficiently than those of Bundibugyo virus (BDBV) and Marburg virus (MARV), resulting in stable replication to facilitate the pathogenesis. Altogether, this study enriches understanding on EBOV evasion of innate immune response, and provides insights into the interplay between filoviruses and host.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , Interferón Tipo I , Humanos , Animales , Proteínas Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/genética , Inmunidad Innata , Ebolavirus/genética , Replicación Viral
16.
Signal Transduct Target Ther ; 8(1): 350, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37709783

RESUMEN

The ongoing pandemic caused by mpox virus (MPXV) has become an international public health emergency that poses a significant threat to global health. The vaccinia virus Tiantan strain (VTT) was used to vaccinate against smallpox in China 42 years ago. It is urgent to assess the level of immunity to smallpox in individuals vaccinated 43 or more years ago and evaluate their immunological susceptibility to MPXV. Here, we recruited 294 volunteers and detected the level of residual humoral immunity, including the vaccinia-specific IgG level and neutralizing antibody titer, and the cross-antibodies of MPXV A29L, B6R, A35R, and M1R. Our results showed that the humoral immunity from the smallpox vaccine in the population still remains, and VTT-specific NAb levels wane with age. The majority of the population pre-1981 who should be immunized with VTT still maintains certain levels of MPXV-specific antibodies, in particular, targeting A35R and B6R antigens. Furthermore, we separately analyzed the correlations between the OD450 values of VTT-specific IgG and A35R-specific IgG, B6R-specific IgG, and A29L-specific IgG with plasma samples diluted 1:40, showing a linear correlation (p < 0.0001). Our findings suggest that most Chinese populations still maintain VTT-specific IgG antibodies for 42 or more years after smallpox vaccination and could provide some level of protection against MPXV.


Asunto(s)
Inmunidad Humoral , Mpox , Vacuna contra Viruela , Humanos , Anticuerpos Neutralizantes , Inmunoglobulina G , Monkeypox virus , Viruela/prevención & control , Vacunación , Vacuna contra Viruela/inmunología , Mpox/prevención & control
17.
MedComm (2020) ; 4(6): e460, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38107058

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron and its subvariants (such as BQ.1, XBB and the latest variants, including XBB.1.16, EG.5, and BA.2.86), as the dominant variants, currently account for almost all new infections in the world due to their high transmissibility and immune escape ability. Omicron-specific mRNA vaccines showed great potential to protect against Omicron infections. However, whether the vaccine could provide long-term protection is unknown. Toward this goal, we evaluated the immunogenicity of a preclinical Omicron (BA.1)-specific mRNA vaccine (SOmicron-6P) in different animal models. SOmicron-6P induced the highest levels of antibody titers at 1-2 weeks in different animals after the second dose. Even 9 months after the immunization, we observed modest neutralizing activity against Omicron subvariants in macaques. In addition, immunological memory cells can be rapidly reactivated upon stimulation. SOmicron-6P at concentrations higher than 10 µg effectively protected hamsters from BA.1 challenge 253 days after the first immunization, which could be attributed to the reactivation of immune systems. In addition, the toxicity tests conducted in rats revealed a highly favorable biosafety profile for SOmicron-6P, even at high dosages. Our data suggest that the Omicron-specific mRNA vaccine is highly effective and safe in animal models and provides long-term immunologic protection against SARS-CoV-2 Omicron infections.

18.
Virol Sin ; 38(1): 119-127, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36450323

RESUMEN

Taurolidine (TRD), a derivative of taurine, has anti-bacterial and anti-tumor effects by chemically reacting with cell-walls, endotoxins and exotoxins to inhibit the adhesion of microorganisms. However, its application in antiviral therapy is seldom reported. Here, we reported that TRD significantly inhibited the replication of influenza virus H5N1 in MDCK cells with the half-maximal inhibitory concentration (EC50) of 34.45 â€‹µg/mL. Furthermore, the drug inhibited the amplification of the cytokine storm effect and improved the survival rate of mice lethal challenged with H5N1 (protection rate was 86%). Moreover, TRD attenuated virus-induced lung damage and reduced virus titers in mice lungs. Administration of TRD reduced the number of neutrophils and increased the number of lymphocytes in the blood of H5N1 virus-infected mice. Importantly, the drug regulated the NF-κB signaling pathway by inhibiting the separation of NF-κB and IκBa, thereby reducing the expression of inflammatory factors. In conclusion, our findings suggested that TRD could act as a potential anti-influenza drug candidate in further clinical studies.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Virus de la Influenza A , Gripe Aviar , Infecciones por Orthomyxoviridae , Animales , Ratones , FN-kappa B/metabolismo , Antivirales/farmacología , Antivirales/uso terapéutico , Infecciones por Orthomyxoviridae/prevención & control , Virus de la Influenza A/fisiología , Transducción de Señal , Taurina/farmacología , Taurina/uso terapéutico , Ratones Endogámicos BALB C , Replicación Viral
19.
Virology ; 585: 145-154, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37348143

RESUMEN

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.


Asunto(s)
COVID-19 , Vacunas , Animales , Ratones , Ratones Endogámicos C57BL , COVID-19/prevención & control , SARS-CoV-2/genética , Adyuvantes Inmunológicos , Glicoproteína de la Espiga del Coronavirus , Anticuerpos Antivirales , Anticuerpos Neutralizantes
20.
Int J Biol Macromol ; 253(Pt 3): 126817, 2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-37690653

RESUMEN

SARS-CoV-2, a type of respiratory virus, has exerted a great impact on global health and economy over the past three years. Antibody-based therapy was initially successful but later failed due to the accumulation of mutations in the spike protein of the virus. Strategies that enable antibodies to resist virus escape are therefore of great significance. Here, we engineer a bispecific SARS-CoV-2 neutralizing nanobody in secretory Immunoglobulin A (SIgA) format, named S2-3-IgA2m2, which shows broad and potent neutralization against SARS-CoV-1, SARS-CoV-2 and its variants of concern (VOCs) including XBB and BQ.1.1. S2-3-IgA2m2 is ∼1800-fold more potent than its parental IgG counterpart in neutralizing XBB. S2-3-IgA2m2 is stable in mouse lungs at least for three days when administrated by nasal delivery. In hamsters infected with BA.5, three intranasal doses of S2-3-IgA2m2 at 1 mg/kg significantly reduce viral RNA loads and completely eliminate infectious particles in the trachea and lungs. Notably, even at single dose of 1 mg/kg, S2-3-IgA2m2 prophylactically administered through the intranasal route drastically reduces airway viral RNA loads and infectious particles. This study provides an effective weapon combating SARS-CoV-2, proposes a new strategy overcoming the virus escape, and lays strategic reserves for rapid response to potential future outbreaks of "SARS-CoV-3".


Asunto(s)
Anticuerpos , SARS-CoV-2 , Animales , Cricetinae , Ratones , Brotes de Enfermedades , Inmunoglobulina A Secretora , ARN Viral , Anticuerpos Antivirales , Anticuerpos Neutralizantes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA