Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Nat Commun ; 14(1): 8440, 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38114531

RESUMEN

Autophagy receptor NDP52 triggers bacterial autophagy against infection. However, the ability of NDP52 to protect against viral infection has not been established. We show that NDP52 binds to envelope proteins of hepatitis B virus (HBV) and triggers a degradation process that promotes HBV clearance. Inactivating NDP52 in hepatocytes results in decreased targeting of viral envelopes in the lysosome and increased levels of viral replication. NDP52 inhibits HBV at both viral entry and late replication stages. In contrast to NDP52-mediated bacterial autophagy, lysosomal degradation of HBV envelopes is independent of galectin 8 and ATG5. NDP52 forms complex with Rab9 and viral envelope proteins and links HBV to Rab9-dependent lysosomal degradation pathway. These findings reveal that NDP52 acts as a sensor for HBV infection, which mediates a unique antiviral response to eliminate the virus. This work also suggests direct roles for autophagy receptors in other lysosomal degradation pathways than canonical autophagy.


Asunto(s)
Virus de la Hepatitis B , Hepatitis B , Humanos , Virus de la Hepatitis B/fisiología , Hepatocitos/metabolismo , Autofagia/fisiología , Lisosomas/metabolismo , Antivirales/farmacología , Antivirales/uso terapéutico , Antivirales/metabolismo , Replicación Viral/fisiología
2.
Emerg Microbes Infect ; 10(1): 1947-1959, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34520320

RESUMEN

Human adenovirus types 4 (HAdV4) and 7 (HAdV7) often lead to severe respiratory diseases and occur epidemically in children, adults, immune deficiency patients, and other groups, leading to mild or severe symptoms and even death. However, no licensed adenovirus vaccine has been approved in the market for general use. E3 genes of adenovirus are generally considered nonessential for virulence and replication; however, a few studies have demonstrated that the products of these genes are also functional. In this study, most of the E3 genes were deleted, and two E3-deleted recombinant adenoviruses (ΔE3-rAdVs) were constructed as components of the vaccine. After E3 deletion, the replication efficiencies and cytopathogenicity of ΔE3-rAdVs were reduced, indicating that ΔE3-rAdVs were attenuated after E3 genes deletion. Furthermore, single immunization with live-attenuated bivalent vaccine candidate protects mice against challenge with wild-type human adenovirus types 4 and 7, respectively. Vaccinated mice demonstrated remarkably decreased viral loads in the lungs and less lung pathology compared to the control animals. Taken together, our study confirms the possibility of the two live-attenuated viruses as a vaccine for clinic use and illustrates a novel strategy for the construction of an adenovirus vaccine.


Asunto(s)
Proteínas E3 de Adenovirus/genética , Infecciones por Adenovirus Humanos/prevención & control , Vacunas contra el Adenovirus/inmunología , Adenovirus Humanos/inmunología , Vacunas Atenuadas/inmunología , Células A549 , Infecciones por Adenovirus Humanos/inmunología , Adenovirus Humanos/clasificación , Animales , Línea Celular , Femenino , Eliminación de Gen , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Carga Viral
3.
Cell Discov ; 7(1): 123, 2021 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-34923570

RESUMEN

A safe and effective vaccine for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is urgently needed to tackle the COVID-19 global pandemic. Here, we describe the development of chimpanzee adenovirus serotypes 6 and 68 (AdC6 and AdC68) vector-based vaccine candidates expressing the full-length transmembrane spike glycoprotein. We assessed the vaccine immunogenicity, protective efficacy, and immune cell profiles using single-cell RNA sequencing in mice. Mice were vaccinated via the intramuscular route with the two vaccine candidates using prime-only regimens or heterologous prime-boost regimens. Both chimpanzee adenovirus-based vaccines elicited strong and long-term antibody and T cell responses, balanced Th1/Th2 cell responses, robust germinal center responses, and provided effective protection against SARS-CoV-2 infection in mouse lungs. Strikingly, we found that heterologous prime-boost immunization induced higher titers of protective antibodies, and more spike-specific memory CD8+ T cells in mice. Potent neutralizing antibodies produced against the highly transmissible SARS-CoV-2 variants B.1.1.7 lineage (also known as N501Y.V1) and B.1.351 lineage (also known as N501Y.V2) were detectable in mouse sera over 6 months after prime immunization. Our results demonstrate that the heterologous prime-boost strategy with chimpanzee adenovirus-based vaccines is promising for further development to prevent SARS-CoV-2 infection.

4.
Front Immunol ; 12: 697074, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34262569

RESUMEN

The development of a safe and effective vaccine against SARS-CoV-2, the causative agent of pandemic coronavirus disease-2019 (COVID-19), is a global priority. Here, we aim to develop novel SARS-CoV-2 vaccines based on a derivative of less commonly used rare adenovirus serotype AdC68 vector. Three vaccine candidates were constructed expressing either the full-length spike (AdC68-19S) or receptor-binding domain (RBD) with two different signal sequences (AdC68-19RBD and AdC68-19RBDs). Single-dose intramuscular immunization induced robust and sustained binding and neutralizing antibody responses in BALB/c mice up to 40 weeks after immunization, with AdC68-19S being superior to AdC68-19RBD and AdC68-19RBDs. Importantly, immunization with AdC68-19S induced protective immunity against high-dose challenge with live SARS-CoV-2 in a golden Syrian hamster model of SARS-CoV-2 infection. Vaccinated animals demonstrated dramatic decreases in viral RNA copies and infectious virus in the lungs, as well as reduced lung pathology compared to the control animals. Similar protective effects were also found in rhesus macaques. Taken together, these results confirm that AdC68-19S can induce protective immune responses in experimental animals, meriting further development toward a human vaccine against SARS-CoV-2.


Asunto(s)
Vacunas contra el Adenovirus/administración & dosificación , Vacunas contra la COVID-19/administración & dosificación , COVID-19/prevención & control , Esquemas de Inmunización , Inmunogenicidad Vacunal , SARS-CoV-2/inmunología , Vacunación/métodos , Vacunas contra el Adenovirus/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , COVID-19/virología , Vacunas contra la COVID-19/inmunología , Cricetinae , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Pan troglodytes , ARN Viral/sangre , Glicoproteína de la Espiga del Coronavirus/inmunología , Transfección , Resultado del Tratamiento
5.
Hum Vaccin Immunother ; 16(10): 2389-2402, 2020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-32078405

RESUMEN

Oncolytic viruses have been taking the front stage in biological therapy for cancer recently. The first and most potent virus to be used in oncolytic virotherapy is human adenovirus. Recently, ongoing extensive research has suggested that other viruses like herpes simplex virus (HSV) and measles virus can also be considered as potential candidates in cancer therapy. An HSV-based oncolytic virus, T-VEC, has completed phase Ш clinical trial and has been approved by the U.S. Food and Drug Administration (FDA) for use in biological cancer therapy. Moreover, the vaccine strain of the measles virus has shown impressive results in pre-clinical and clinical trials. Considering their therapeutic efficacy, safety, and reduced side effects, the use of such engineered viruses in biological cancer therapy has the potential to establish a milestone in cancer research. In this review, we summarize the recent clinical advances in the use of oncolytic viruses in biological therapy for cancer. Additionally, this review evaluates the potential viral candidates for their benefits and shortcomings and sheds light on the future prospects.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Humanos , Neoplasias/terapia , Virus Oncolíticos/genética
6.
NPJ Vaccines ; 5(1): 57, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32665862

RESUMEN

Due to the high mutation and recombination rates of the influenza virus, current clinically licensed influenza vaccines and anti-influenza drugs provide limited protection against the emerging influenza virus epidemic. Therefore, universal influenza vaccines with high efficacy are urgently needed to ensure human safety and health. Passive immunization of influenza broadly neutralizing antibodies may become an ideal option for controlling influenza infection. CR9114 isolated from the peripheral blood mononuclear cells of healthy donors is a broadly neutralizing monoclonal antibody that targets different types of influenza viruses. As the adenovirus vector is one of the most promising delivery vehicles, we employed the chimpanzee adenoviral vector, AdC68, to express CR9114 as a universal anti-influenza vaccine, termed AdC68-CR9114, and evaluated its antibody expression and its broad spectrum of prophylactic and therapeutic effects in animal models. Based on our findings, AdC68-CR9114-infected cell expressed the broadly neutralizing antibody at a high level in vitro and in vivo, exhibited biological functions, and protected mice from different types of influenza virus infection at different time points. The findings from this study shed light on a new strategy for controlling and preventing influenza infection.

7.
Hum Vaccin Immunother ; 14(7): 1679-1685, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29300685

RESUMEN

Adenoviral vector has been employed as one of the most efficient means against infectious diseases and cancer. It can be genetically modified and armed with foreign antigens to elicit specific antibody responses and T cell responses in hosts as well as engineered to induce apoptosis in cancer cells. The chimpanzee adenovirus-based vector is one kind of novel vaccine carriers whose unique features and non-reactivity to pre-existing human adenovirus neutralizing antibodies makes it an outstanding candidate for vaccine research and development. Here, we review the different strategies for constructing chimpanzee adenoviral vectors and their applications in recent clinical trials and also discuss the oncolytic virotherapy and immunotherapy based on chimpanzee adenoviral vectors.


Asunto(s)
Adenovirus de los Simios/genética , Vectores Genéticos , Vacunas/genética , Adenovirus Humanos/genética , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Ensayos Clínicos como Asunto , Humanos , Inmunoterapia , Ratones , Viroterapia Oncolítica , Pan troglodytes/virología , Linfocitos T/inmunología , Vacunas/inmunología
8.
Exp Toxicol Pathol ; 68(4): 223-31, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26775023

RESUMEN

Atrazine (ATR) is primarily distributed in liver and hazardous to animal health. Cytochrome P450 enzyme system (CYP450s) is responsible for the biotransformation of toxic substances. Lycopene (LYC) prevents the herbicide-induced toxicity. However, it is unclear that LYC protects against ATR-induced hepatotoxicity via modifying CYP450s. To ascertain the chemoprevention of LYC on ATR-induced hepatotoxicity, male Kunming mice were treated with LYC (5mg/kg) and/or ATR (50mg/kg or 200mg/kg) by gavage administration for 21 days. These results showed that ATR induced the increase of total CYP450 and Cytochrome b5 (Cyt b5) contents and stimulated the activities of CYP450s enzymes (erythromycin N-demethylase (ERND), aminopyrin N-demethylase (APND), aniline-4-hydeoxylase (AH) and NADPH-cytochrome c reductase (NCR)) in hepatic microsomes. The mRNA expressions of six CYP450s genes (increase: CYP1a1, CYP2a4, CYP3a57 and decrease: CYP2f2, CYP3a11, CYP4a31) were significantly influenced by ATR. LYC modulated the contents and activities of CYP450s and normalized the expressions of four CYP450s genes (CYP1b1, CYP2a4, CYP2e1, and 4A14). These findings suggested that ATR induced hepatic CYP450s disturbance and influenced the gene expression of CYP450s. Lycopene protected against hepatic CYP450s disturbance induced by ATR via modifying the hepatic CYP450s activities and transcription in mice.


Asunto(s)
Antioxidantes/uso terapéutico , Atrazina/toxicidad , Carotenoides/uso terapéutico , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Sistema Enzimático del Citocromo P-450/metabolismo , Contaminantes Ambientales/toxicidad , Microsomas Hepáticos/efectos de los fármacos , Animales , Antioxidantes/administración & dosificación , Biomarcadores/metabolismo , Carotenoides/administración & dosificación , Enfermedad Hepática Inducida por Sustancias y Drogas/enzimología , Relación Dosis-Respuesta a Droga , Licopeno , Masculino , Ratones Endogámicos , Microsomas Hepáticos/enzimología , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Environ Pollut ; 207: 299-307, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26432752

RESUMEN

There has been a gradual increase in production and consumption of atrazine (ATR) in agriculture to meet the population rising demands. Female reproduction is necessary for growth and maintenance of population. However, ATR impact on females and particularly ovarian developmental toxicity is less clear. The aim of this study was to define the pathways by which ATR exerted toxic effects on ovarian development of ovary and hypothalamo-pituitary-ovarian (HPO) axis. Female quails were dosed by oral gavage from sexual immaturity to maturity with 0, 50, 250 and 500 mg ATR/kg/d for 45 days. ATR had no effect on mortality but depressed feed intake and growth and influenced the biochemical parameters. Notably, the arrested development of ovaries and oviducts were observed in ATR-exposed quails. The circulating concentrations of E2, P, LH and PRL were unregulated and FSH and T was downregulated in ATR-treated quails. The mRNA expression of GnRH in hypothalamo and LH in pituitary and FSH in ovary was downregulated significantly by ATR exposure and FSH and PRL in pituitary were upregulated. ATR exposure upregulated the level of P450scc, P450arom, 3ß-HSD and 17ß-HSD in ovary and downregulated ERß expression in female quails. However, ATR did not change ERα expression in ovary. This study provides new insights regarding female productive toxicology of ATR exposure. Ovary and oviduct in sexually maturing females were target organs of ATR-induced developmental toxicity. We propose that ATR-induced developmental abnormality of ovary and oviduct is associated with disruption of gonadal hormone balance and HPO axis in female quails.


Asunto(s)
Atrazina/toxicidad , Coturnix/crecimiento & desarrollo , Ovario/efectos de los fármacos , Oviductos/efectos de los fármacos , Animales , Peso Corporal , Femenino , Hormonas Gonadales , Hormona Liberadora de Gonadotropina/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Ovario/crecimiento & desarrollo , Oviductos/crecimiento & desarrollo , Hipófisis/efectos de los fármacos , Reproducción/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA