Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Intervalo de año de publicación
1.
Eur J Med Chem ; 276: 116694, 2024 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-39047607

RESUMEN

As a highly conserved signaling network across different species, the Hippo pathway is involved in various biological processes. Dysregulation of the Hippo pathway could lead to a wide range of diseases, particularly cancers. Extensive researches have demonstrated the close association between dysregulated Hippo signaling and tumorigenesis as well as tumor progression. Consequently, targeting the Hippo pathway has emerged as a promising strategy for cancer treatment. In fact, there has been an increasing number of reports on small molecules that target the Hippo pathway, exhibiting therapeutic potential as anticancer agents. Importantly, some of Hippo signaling pathway inhibitors have been approved for the clinical trials. In this work, we try to provide an overview of the core components and signal transduction mechanisms of the Hippo signaling pathway. Furthermore, we also analyze the relationship between Hippo signaling pathway and cancers, as well as summarize the small molecules with proven anti-tumor effects in clinical trials or reported in literatures. Additionally, we discuss the anti-tumor potency and structure-activity relationship of the small molecule compounds, providing a valuable insight for further development of anticancer agents against this pathway.

2.
Bioorg Med Chem ; 107: 117762, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38759254

RESUMEN

Honokiol, derived from Magnolia officinalis (a traditional Chinese medicine), has been reported to have anticancer activity. Here, a series of novel honokiol thioethers bearing a 1,3,4-oxadiazole moiety were prepared and evaluated for their anticancer activities against three types of digestive system tumor cells. Biological evaluation showed that honokiol derivative 3k exhibited the best antiproliferative activity against HCT116 cells with an IC50 value of 6.1 µmol/L, superior to the reference drug 5-fluorouracil (IC50: 9.63 ± 0.27 µmol/L). The structure-activity relationships (SARs) indicated that the introduction of -(4-NO2)Ph, 3-pyridyl, -(2-F)Ph, -(4-F)Ph, -(3-F)Ph, -(4-Cl)Ph, and -(3-Cl)Ph groups was favorable for enhancing the anticancer activity of the title honokiol thioethers. Further study revealed that honokiol thioether 3k can well inhibit the proliferation of colon cancer cells HCT116, arresting the cells in G1 phase and inducing cell death. Moreover, a preliminary mechanism study indicated that 3k directly inhibits the transcription and expression of YAP protein without activating the Hippo signaling pathway. Thus, honokiol thioether 3k could be deeply developed for the development of honokiol-based anticancer candidates.


Asunto(s)
Compuestos de Bifenilo , Proliferación Celular , Ensayos de Selección de Medicamentos Antitumorales , Lignanos , Proteínas Señalizadoras YAP , Humanos , Lignanos/farmacología , Lignanos/química , Lignanos/síntesis química , Compuestos de Bifenilo/farmacología , Compuestos de Bifenilo/antagonistas & inhibidores , Compuestos de Bifenilo/química , Relación Estructura-Actividad , Proliferación Celular/efectos de los fármacos , Células HCT116 , Proteínas Señalizadoras YAP/metabolismo , Estructura Molecular , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Neoplasias del Colon/metabolismo , Sulfuros/química , Sulfuros/farmacología , Sulfuros/síntesis química , Factores de Transcripción/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Antineoplásicos Fitogénicos/farmacología , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/síntesis química , Relación Dosis-Respuesta a Droga , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Compuestos Alílicos , Fenoles
3.
Phytomedicine ; 118: 154943, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37421765

RESUMEN

BACKGROUND: Shikonin, a natural naphthoquinone compound, has a wide range of pharmacological effects, but its anti-tumor effect and underlying mechanisms in bladder cancer remain unclear. PURPOSE: We aimed to investigate the role of shikonin in bladder cancer in vitro and in vivo in order to broaden the scope of shikonin's clinical application. STUDY DESIGN AND METHODS: We performed MTT and colony formation to detect the inhibiting effect of shikonin on bladder cancer cells. ROS staining and flow cytometry assays were performed to detect the accumulation of ROS. Western blotting, siRNA and immunoprecipitation were used to evaluate the effect of necroptosis in bladder cancer cells. Transmission electron microscopy and immunofluorescence were used to examine the effect of autophagy. Nucleoplasmic separation and other pharmacological experimental methods described were used to explore the Nrf2 signal pathway and the crosstalk with necroptosis and autophagy. We established a subcutaneously implanted tumor model and performed immunohistochemistry assays to study the effects and the underlying mechanisms of shikonin on bladder cancer cells in vivo. RESULTS: The results showed that shikonin has a selective inhibitory effect on bladder cancer cells and has no toxicity on normal bladder epithelial cells. Mechanically, shikonin induced necroptosis and impaired autophagic flux via ROS generation. The accumulation of autophagic biomarker p62 elevated p62/Keap1 complex and activated the Nrf2 signaling pathway to fight against ROS. Furthermore, crosstalk between necroptosis and autophagy was present, we found that RIP3 may be involved in autophagosomes and be degraded by autolysosomes. We found for the first time that shikonin-induced activation of RIP3 may disturb the autophagic flux, and inhibiting RIP3 and necroptosis could accelerate the conversion of autophagosome to autolysosome and further activate autophagy. Therefore, on the basis of RIP3/p62/Keap1 complex regulatory system, we further combined shikonin with late autophagy inhibitor(chloroquine) to treat bladder cancer and achieved a better inhibitory effect. CONCLUSION: In conclusion, shikonin could induce necroptosis and impaired autophagic flux through RIP3/p62/Keap1 complex regulatory system, necroptosis could inhibit the process of autophagy via RIP3. Combining shikonin with late autophagy inhibitor could further activate necroptosis via disturbing RIP3 degradation in bladder cancer in vitro and in vivo.


Asunto(s)
Naftoquinonas , Neoplasias de la Vejiga Urinaria , Humanos , Especies Reactivas de Oxígeno/metabolismo , Necroptosis , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Muerte Celular , Naftoquinonas/farmacología , Autofagia , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico
4.
J Enzyme Inhib Med Chem ; 38(1): 2237701, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37489043

RESUMEN

In this work, a series of novel arylamide derivatives containing piperazine moiety were designed and synthesised as tubulin polymerisation inhibitors. Among 25 target compounds, compound 16f (MY-1121) exhibited low nanomolar IC50 values ranging from 0.089 to 0.238 µM against nine human cancer cells. Its inhibitory effects on liver cancer cells were particularly evident with IC50 values of 89.42 and 91.62 nM for SMMC-7721 and HuH-7 cells, respectively. Further mechanism studies demonstrated that compound 16f (MY-1121) could bind to the colchicine binding site of ß-tubulin and directly act on ß-tubulin, thus inhibiting tubulin polymerisation. Additionally, compound 16f (MY-1121) could inhibit colony forming ability, cause morphological changes, block cell cycle arrest at the G2 phase, induce cell apoptosis, and regulate the expression of cell cycle and cell apoptosis related proteins in liver cancer cells. Overall, the promising bioactivities of compound 16f (MY-1121) make the novel arylamide derivatives have the value for further development as tubulin polymerisation inhibitors with potent anticancer activities.


Asunto(s)
Neoplasias Hepáticas , Tubulina (Proteína) , Humanos , Apoptosis , Sitios de Unión , Piperazina , Moduladores de Tubulina
5.
Bioorg Chem ; 137: 106580, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37149948

RESUMEN

As a class of microtubule targeting agents, colchicine binding site inhibitors (CBSIs) are considered as promising drug candidates for cancer therapy. However, due to adverse reactions, there are currently no CBSIs approved by FDA for cancer treatment. Therefore, extensive efforts are still encouraged to find novel CBSIs with different chemical structures and better anticancer efficacies. In this work, we designed and synthesized a new coumarin-dihydroquinoxalone derivative, MY-673, and evaluated its anticancer potency in vitro and in vivo. We confirmed that MY-673 was a potent CBSI that it not only inhibited tubulin polymerization, but also exhibited significant inhibitory potency on the growth of 13 cancer cells with IC50 values from 11.7 nM to 395.9 nM. Based on the results of kinase panel screening, MY-673 could inhibit ERK (extracellular regulated protein kinases) pathways-related kinases. We further confirmed that MY-673 could inhibit ERK signaling pathway in MGC-803 and HGC-27 cells, and then affected the expression level of SMAD4 protein in TGF-ß (transforming growth factor ß) /SMAD (small mother against decapentaplegic) signaling pathway using the western blotting assay. In addition, compound MY-673 could effectively inhibit cell proliferation, migration and induce cell apoptosis. We also further confirmed the in vivo efficacy of MY-673 in inhibiting tumor growth using the MGC-803 xenograft tumor model. At 20 mg/kg, the TGI rate was 85.9%, and it did not cause obvious toxicity to the main organs of mice. Together, the results we report here indicated that MY-673 was a promising CBSI for cancer treatment, which was capable of inhibiting the ERK pathway with potent antiproliferative activities in vitro and in vivo.


Asunto(s)
Antineoplásicos , Neoplasias Gástricas , Humanos , Animales , Ratones , Moduladores de Tubulina/farmacología , Moduladores de Tubulina/uso terapéutico , Moduladores de Tubulina/química , Sistema de Señalización de MAP Quinasas , Tubulina (Proteína)/metabolismo , Microtúbulos , Colchicina/metabolismo , Proliferación Celular , Neoplasias Gástricas/tratamiento farmacológico , Antineoplásicos/química , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Relación Estructura-Actividad
6.
Eur J Med Chem ; 252: 115281, 2023 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-36940611

RESUMEN

In this work, N-benzylarylamide-dithiocarbamate based derivatives were designed, synthesized, and their biological activities as anticancer agents were explored. Some of the 33 target compounds displayed significant antiproliferative activities with IC50 values at the double-digit nanomolar level. The representative compound I-25 (also named MY-943) not only showed the most effective inhibitory effects on three selected cancer cells MGC-803 (IC50 = 0.017 µM), HCT-116 (IC50 = 0.044 µM) and KYSE450 (IC50 = 0.030 µM), but also exhibited low nanomolar IC50 values from 0.019 to 0.253 µM against the other 11 cancer cells. Compound I-25 (MY-943) effectively inhibited tubulin polymerization and suppressed LSD1 at the enzymatic levels. Compound I-25 (MY-943) could act on the colchicine binding site of ß-tubulin, thus disrupting the construction of cell microtubule network and affecting the mitosis. In addition, compound I-25 (MY-943) could dose-dependently induce the accumulation of H3K4me1/2 (MGC-803 and SGC-7091 cells) and H3K9me2 (SGC-7091 cells). Compound I-25 (MY-943) could induce G2/M phase arrest and cell apoptosis, and suppress migration in MGC-803 and SGC-7901 cells. In addition, compound I-25 (MY-943) significantly modulated the expression of apoptosis- and cycle-related proteins. Furthermore, the binding modes of compound I-25 (MY-943) with tubulin and LSD1 were explored by molecular docking. The results of in vivo anti-gastric cancer assays using in situ tumor models showed that compound I-25 (MY-943) effectively reduced the weight and volume of gastric cancer in vivo without obvious toxicity. All these findings suggested that the N-benzylarylamide-dithiocarbamate based derivative I-25 (MY-943) was an effective dual inhibitor of tubulin polymerization and LSD1 that inhibited gastric cancers.


Asunto(s)
Antineoplásicos , Neoplasias Gástricas , Humanos , Tubulina (Proteína)/metabolismo , Línea Celular Tumoral , Simulación del Acoplamiento Molecular , Polimerizacion , Proliferación Celular , Moduladores de Tubulina/farmacología , Moduladores de Tubulina/química , Antineoplásicos/farmacología , Antineoplásicos/química , Neoplasias Gástricas/tratamiento farmacológico , Histona Demetilasas/metabolismo , Relación Estructura-Actividad , Ensayos de Selección de Medicamentos Antitumorales
7.
Eur J Med Chem ; 240: 114583, 2022 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-35834904

RESUMEN

Novel N-benzylarylamide saderivatives were designed and synthesized, and their antiproliferative activities were explored. Some of 51 target compounds exhibited potent inhibitory activities against MGC-803, HCT-116 and KYSE450 cells with IC50 values in two-digit nanomolar. Compound I-33 (MY-875) displayed the most potent antiproliferative activities against MGC-803, HCT-116 and KYSE450 cells (IC50 = 0.027, 0.055 and 0.067 µM, respectively) and possessed IC50 values ranging from 0.025 to 0.094 µM against other 11 cancer cell lines. Further mechanism studies indicated that compound I-33 (MY-875) inhibited tubulin polymerization (IC50 = 0.92 µM) by targeting the colchicine bingding site of tubulin. Compound I-33 (MY-875) disrupted the construction of the microtubule networks and affected the mitosis in MGC-803 and SGC-7901 cells. In addition, although it acted as a colchicine binding site inhibitor, compound I-33 (MY-875) also activated the Hippo pathway to promote the phosphorylation status of MST and LATS, resulting in the YAP degradation in MGC-803 and SGC-7901 cells. Due to the degradation of YAP, the expression levels of TAZ and Axl decreased. Because of the dual actions on colchicine binding site and Hippo pathway, compound I-33 (MY-875) dose-dependently inhibited cell colony formatting ability, arrested cells at the G2/M phase and induced cells apoptosis in MGC-803 and SGC-7901 cells. Moreover, compound I-33 (MY-875) could regulate the levels of cell cycle and apoptosis regulatory proteins in MGC-803 and SGC-7901 cells. Furthermore, molecular docking analysis suggested that the hydrogen bond and hydrophobic interactions made compound I-33 (MY-875) well bind into the colchicine binding site of tubulin. Collectively, compound I-33 (MY-875) is a novel anti-gastric cancer agent and deserves to be further investigated for cancer therapy by targeting the colchicine binding site of tubulin and activating the Hippo pathway.


Asunto(s)
Antineoplásicos , Moduladores de Tubulina , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular , Colchicina/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Vía de Señalización Hippo , Simulación del Acoplamiento Molecular , Polimerizacion , Relación Estructura-Actividad , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacología
8.
Eur J Med Chem ; 238: 114467, 2022 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-35605363

RESUMEN

Novel coumarin-indole derivatives were designed, synthesized and evaluated as tubulin polymerization inhibitors targeting the colchicine binding site. Among these compounds, compound MY-413 displayed the most potent inhibitory activities against gastric cancer cell line MGC-803 with an IC50 value of 0.011 µM. Furthermore, the IC50 values of compound MY-413 was less than 0.1 µM for other 17 cancer cell lines and less than 0.05 µM for other 8 cancer cell lines. Compound MY-413 effectively inhibited the tubulin polymerization (IC50 = 2.46 µM) by binding to the colchicine site. Screening for the inhibitory effects of compound MY-413 on 61 kinases, it was found that compound MY-413 could inhibit MAPK pathways-related kinases. Because of the inhibitory effects of compound MY-413 on tubulin polymerization and MAPK signaling pathway, compound MY-413 induced cell apoptosis, arrested the cell cycle in the G2/M phase, induced the inhibition of cell proliferation and migration in gastric cancer cells MGC-803 and HGC-27. In addition, compound MY-413 could significantly inhibit tumor growth in MGC-803 xenograft tumor models with tumor growth inhibition (TGI) rates of 70% (15 mg/kg) and 80% (30 mg/kg) without obvious toxicity. Consistent with the in vitro results, compound MY-413 also inhibited MAPK signaling pathway, and induced apoptosis and proliferation inhibition in vivo. In conclusion, this work indicated that compound MY-413 was a promising lead compound for the further investigation as a potential anti-gastric cancer agent.


Asunto(s)
Antineoplásicos , Neoplasias Gástricas , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular , Colchicina/farmacología , Cumarinas/farmacología , Cumarinas/uso terapéutico , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Indoles/química , Indoles/farmacología , Polimerizacion , Neoplasias Gástricas/tratamiento farmacológico , Relación Estructura-Actividad , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/química
9.
Biochem Pharmacol ; 201: 115070, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35526597

RESUMEN

Given the essential role of Epigenetic regulation in many biological processes, targeted epigenetic drugs have been gradually applied to the treatment of tumors. Histone deacetylases (HDACs) are a class of epigenetic enzymes, which play key roles in chromosome structural modification and gene expression regulation. Targeted microtubules drugs have achieved great success in clinical application for decades. Development of novel agents with multitargeting capabilities specially dual-target has become a popular research field for the treatment of human cancers, which may provide synergistic anticancer effects. Here, we reported a novel aromatic amide derivative SY-65 co-targeted tubulin and histone deacetylase 1 with potent anticancer activity in vitro and in vivo. Compound SY-65 was identified as a dual inhibitor of tubulin/HDAC1 (IC50 = 3.64 and 0.529 µM, respectively) with excellent antiproliferative activity against MGC-803, HCT-116, KYSE-450, HGC-27, SGC-7901 and MKN-45 cells. Especially, compound SY-65 exhibited potent antiproliferative activity against MGC-803, HGC-27 and SGC-7901 cells with IC50 values <55 nM, which was better than that of Colchicine, MS-275 and SAHA. Compound SY-65 effectively inhibited tubulin polymerization and bound to the colchicine binding site of tubulin, as well as inhibited HDAC1 activity both intra/extracellularly. Molecular docking results suggested there were the well-defined binding modes of compound SY-65 in HDAC1 and tubulin. In addition, compound SY-65 inhibited colony formation, interfered with the cell cycle distribution, induced cell cycle arrest at the G2/M phase and apoptosis in MGC-803 and HGC-27 cells. Compound SY-65 also exhibited a good tumor inhibitory effect in vivo without obvious toxicity. Therefore, compound SY-65 could be developed as a novel tubulin/HDAC1 candidate inhibitor for future cancer therapeutics.


Asunto(s)
Antineoplásicos , Neoplasias , Amidas/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular , Colchicina/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Epigénesis Genética , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 1/farmacología , Humanos , Simulación del Acoplamiento Molecular , Neoplasias/tratamiento farmacológico , Relación Estructura-Actividad , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/farmacología
10.
Eur J Med Chem ; 225: 113801, 2021 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-34455358

RESUMEN

NEDDylation process regulates multiple physiological functions and signaling pathways, which are still in an equilibrium that favors the survival and proliferation of tumor cells. Unlike inhibitors, NEDDylation agonists are rarely studied. In this work, novel 1,2,4-triazine-dithiocarbamate derivatives were synthesized and evaluated for antiproliferative activity against MGC-803, PC-3 and EC-109 cells. Among them, compound K3 displayed the most potent activity MGC-803, PC-3 and EC-109 cells with IC50 values of 2.35, 5.71 and 10.1 µM, respectively, which were more potent than 5-FU. Further cellular mechanisms suggested that compound K3 inhibited the cell viability, induced proliferation inhibition, arrested cell cycle at G2/M phase and induced cell apoptosis in MGC-803 and HGC-27 cells. Importantly, compound K3 could interact with NAE1 to promote the NEDDylation of MGC-803 and HGC-27 cells. The promotion of NEDDylation resulted in the degradation of c-IAP and YAP/TAZ, which leads to the induction of cell apoptosis and inhibition of proliferation in MGC-803 and HGC-27 cells. Therefore, as a NEDDylation agonist, compound K3 could effectively inhibit gastric cancer cells. Here, we reported NEDDylation promotion induced by compound K3, which could inhibit the cancer cell lines MGC-803 and HGC-27 and induce the cancer cell apoptosis via prompting the degradation of c-IAP and YAP/TAZ.


Asunto(s)
Antineoplásicos/farmacología , Descubrimiento de Drogas , Neoplasias Gástricas/tratamiento farmacológico , Triazinas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Estructura Molecular , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Relación Estructura-Actividad , Triazinas/síntesis química , Triazinas/química , Células Tumorales Cultivadas
11.
J Control Release ; 319: 438-449, 2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-31926191

RESUMEN

Metronomic cancer chemotherapy has displayed the potential to ameliorate immunosuppressive tumor microenvironment (TME) and facilitate antitumor immunotherapy, but this strategy requires uninterrupted administration of low-dose chemotherapeutic agents and suffers from rapid drug clearance. Here, we developed a single-dose in situ immune stimulator storage to achieve prolonged retention and sustained release of drugs in tumor parenchyma. Importantly, this storage could initiate immune responses through photothermal therapy (PTT) and simultaneously remodel TME. In detail, the storage framework (NGOPC) with size of ~60 nm, was composed of Ala-Ala-Asn-Cys-Lys modified nano graphene oxide (NGO-PEG-pep) and 2-cyano-6-aminobenzothiazole modified NGO (NGO-PEG-CABT), and could sufficiently penetrate into deep tumor region. Once NGOPC arrived at the core field, legumain overexpressing in TME could trigger click cycloaddition reaction of NGO-PEG-pep with NGO-PEG-CABT to form network, leading to aggregation and augmented retention in tumor. Additionally, paclitaxel (PTX) that can block immunologic escape was loaded in NGOPC (NGOPC@PTX), which synergistically worked with PTT-generated antitumor immunity. We found that NGOPC@PTX possessed the superior ability to accumulate in tumor and generate antitumor immunological efficacy by improving immune factors: induction of HSP70-mediated immunogenic cell death, reduction of regulatory T cells, and activation of cytotoxic T lymphocyte. This in situ storage, which exhibited excellent tumor growth inhibition effect and prolonged lifespan in combination with PTT, displays the potential for intensified cancer immunotherapy.


Asunto(s)
Paclitaxel , Línea Celular Tumoral
12.
Virus Genes ; 48(1): 48-55, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24085475

RESUMEN

This study was conducted to evaluate the virulence and evolution of genotype IX Newcastle disease virus (NDV) isolates obtained from wild birds in the northern Qinling Mountains of China. Five isolates were obtained from 374 larynx and cloacae swabs, which were collected from multiple asymptomatic wild bird species from August 2008 to July 2011, and were subsequently characterized by pathotype and genotype. Deduced amino acid sequences revealed that all five NDV isolates exhibited velogenic fusion protein cleavage sites motif (112)R-R-Q-R-R-F(117), shared as high as 99.8-99.9 % homology with each other, and varied in pathotype by intracerebral pathogenicity indices (ICPI) of 0.425-1.638. Phylogenetic analysis showed that all five isolates were clustered to genotype IX NDV. This is the first study to confirm multiple asymptomatic wild bird species as natural carriers of virulent genotype IX NDV. A novel NDV isolate from the Spotted-necked Dove (family Columbidae) exhibited discordance between its lentogenic ICPI and its virulent proteolytic cleavage site motif (112)R-R-Q-R-R-F(117). Although the five isolates underwent several amino acid mutations in the fusion protein, evidence of continuous evolutionary divergence did exist in the genotype IX NDV, which was always regarded as a conservative genotype.


Asunto(s)
Aves/virología , Portador Sano/veterinaria , Virus de la Enfermedad de Newcastle/clasificación , Virus de la Enfermedad de Newcastle/aislamiento & purificación , Secuencias de Aminoácidos , Animales , Encéfalo/virología , Portador Sano/virología , China , Cloaca/virología , Análisis por Conglomerados , Evolución Molecular , Variación Genética , Genotipo , Laringe/virología , Datos de Secuencia Molecular , Virus de la Enfermedad de Newcastle/genética , Virus de la Enfermedad de Newcastle/patogenicidad , Filogenia , ARN Viral/genética , Análisis de Secuencia de ADN , Homología de Secuencia , Proteínas Virales de Fusión/genética , Virulencia
13.
Virus Genes ; 46(2): 264-70, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23143787

RESUMEN

This paper describes the complete genome sequence of HX01, an isolate of the Newcastle disease virus (NDV) collected from a swine disease outbreak. The genome is 15,186 nt long and consists of six genes in the order of 3'-NP-P-M-F-HN-L-5'. This genome has the same length as the old NDV genotypes (I-IV), whereas the new NDV genotypes (V-IX) are 15,192 nt long. Compared with the genomic sequences of the reference NDV strains, the HX01 genome is highly similar to the genome of other NDV strains. However, some unique features of the HN gene were found in HX01. HX01 possesses the motif (112)G-R-Q-G-R-L(117) at the fusion protein cleavage site, which is typical of lentogenic strains. Pathogenicity tests based on the mean death time and the intracerebral pathogenicity index also revealed the isolate's lentogenic character. Phylogenetic analysis based on the variable region of the F gene (nt 47-420) revealed that HX01 was clustered to genotype II within class II NDV. Genetically, HX01 has a high similarity with the La Sota vaccine strain based on the single gene or complete genomic but is far different from the prevalent genotype VIId NDV which circulates in fowls and waterfowls in mainland China.


Asunto(s)
Enfermedad de Newcastle/virología , Virus de la Enfermedad de Newcastle/genética , Virus de la Enfermedad de Newcastle/aislamiento & purificación , Enfermedades de los Porcinos/virología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Embrión de Pollo , China , Genoma Viral , Genómica , Datos de Secuencia Molecular , Virus de la Enfermedad de Newcastle/química , Virus de la Enfermedad de Newcastle/clasificación , Filogenia , Alineación de Secuencia , Porcinos , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/genética
14.
Vet Immunol Immunopathol ; 145(1-2): 277-82, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22154735

RESUMEN

Vaccine antigens for rabbit hemorrhagic disease virus (RHDV) are currently derived from inactivated RHDV obtained from the livers of experimentally infected rabbits or from several recombinant immunogens. However, the application of these vaccine antigens has been restricted because of biosecurity and immunity characteristics. In the current study, a recombinant adenovirus expressing the RHDV capsid protein (VP60) was constructed and the expression of the recombinant protein was identified through western blot analysis using RHDV-positive rabbit sera. Eighteen rabbits were immunized by injection, direct oral instillation, or using bait. They were challenged with RHDV isolate three weeks after boost immunization. In all cases, the rabbits immunized with the recombinant adenovirus developed RHDV-specific antibodies and cell immune response. The rabbits injected with the recombinant adenovirus were completely protected against RHDV challenge. The adenovirus expression system may provide a strategy for the immunization of rabbits, particularly for the control of RHDV in wild rabbits.


Asunto(s)
Infecciones por Caliciviridae/veterinaria , Virus de la Enfermedad Hemorrágica del Conejo , Vacunas Virales/uso terapéutico , Adenoviridae , Administración Oral , Animales , Infecciones por Caliciviridae/inmunología , Infecciones por Caliciviridae/prevención & control , Proteínas de la Cápside/inmunología , Pruebas de Inhibición de Hemaglutinación/veterinaria , Pruebas de Hemaglutinación/veterinaria , Virus de la Enfermedad Hemorrágica del Conejo/inmunología , Inmunidad Celular/inmunología , Inmunidad Humoral , Conejos/inmunología , Conejos/virología , Vacunas Sintéticas , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA