Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Fitoterapia ; 172: 105716, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37926399

RESUMO

Four previously undescribed angucyclinones umezawaones A-D (1-4) were isolated from the liquid cultures of Umezawaea beigongshangensis. Their structures were determined by spectroscopic analyses, single crystal X-ray diffraction, quantum chemical 13C NMR and electronic circular dichroism calculations. All compounds displayed strong inhibitory activities against indoleamine 2,3-dioxygenase and tryptophan-2,3-dioxygenase in enzymatic assay, especially compound 2.


Assuntos
Actinobacteria , Triptofano Oxigenase , Triptofano Oxigenase/química , Triptofano Oxigenase/metabolismo , Anguciclinas e Anguciclinonas , Actinomyces/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase , Estrutura Molecular
2.
Org Lett ; 25(37): 6796-6801, 2023 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-37676817

RESUMO

A sustainable pathway for the synthesis of tetracyclic purinium salts via ruthenium-catalyzed electro-oxidative annulation of C6-arylpurine nucleosides with alkynes without a stoichiometric metal oxidant has been developed. The protocol described herein exhibits high regioselectivity, broad scope, and wide functional group tolerance, allowing efficient coupling of various biologically important molecules including acyclic, ribosyl, arabinosyl, and deoxyribosyl purine nucleoside derivatives. A novel purinoisoquinolinium-coordinated ruthenium(0) sandwich intermediate has been isolated, crystallographically characterized, and electrochemically analyzed, offering direct mechanistic insight.

3.
J Vet Diagn Invest ; 35(2): 168-172, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36600502

RESUMO

Rhabdomyosarcoma (RMS), a malignant mesenchymal neoplasm derived from skeletal muscle, is relatively rare in both human and veterinary medicine. Here we report an unusual case of invasive spindle-cell RMS (SCRMS) with bone infiltration and pathologic fracture in a 3.5-y-old intact female Bulldog. Radiographically, a large, predominantly osteolytic mass in the tibia and fibula of the left hindlimb had features typical of a malignant primary bone tumor. Clinically, osteosarcoma was suspected, and the leg was amputated. Histologically, the mass was composed of loosely interwoven spindle-cell fascicles; tumor cells were fusiform with cigar-shaped nuclei and abundant eosinophilic cytoplasm. The neoplastic cells were strongly immunopositive for vimentin, muscle-specific actin, desmin, myogenin, and myoD1. Invasive SCRMS with osteolysis was diagnosed based on the histologic examination and immunohistochemical (IHC) stains. The dog was alive without any evidence of local recurrence or distant metastasis 18 mo post-surgery. RMS should be included in the differential diagnosis when osteolysis occurs; IHC staining confirmation is of great value for definitive diagnosis and treatment planning.


Assuntos
Neoplasias Ósseas , Doenças do Cão , Osteólise , Osteossarcoma , Rabdomiossarcoma , Sarcoma , Animais , Cães , Feminino , Neoplasias Ósseas/veterinária , Doenças do Cão/diagnóstico , Osteólise/veterinária , Osteossarcoma/diagnóstico , Osteossarcoma/veterinária , Rabdomiossarcoma/diagnóstico , Rabdomiossarcoma/patologia , Rabdomiossarcoma/veterinária , Sarcoma/patologia , Sarcoma/veterinária
4.
PLoS Pathog ; 18(6): e1010667, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35759516

RESUMO

Porcine hemagglutinating encephalomyelitis virus (PHEV) is a highly neurotropic coronavirus belonging to the genus Betacoronavirus. Similar to pathogenic coronaviruses to which humans are susceptible, such as SARS-CoV-2, PHEV is transmitted primarily through respiratory droplets and close contact, entering the central nervous system (CNS) from the peripheral nerves at the site of initial infection. However, the neuroinvasion route of PHEV are poorly understood. Here, we found that BALB/c mice are susceptible to intranasal PHEV infection and showed distinct neurological manifestations. The behavioral study and histopathological examination revealed that PHEV attacks neurons in the CNS and causes significant smell and taste dysfunction in mice. By tracking neuroinvasion, we identified that PHEV invades the CNS via the olfactory nerve and trigeminal nerve located in the nasal cavity, and olfactory sensory neurons (OSNs) were susceptible to viral infection. Immunofluorescence staining and ultrastructural observations revealed that viral materials traveling along axons, suggesting axonal transport may engage in rapid viral transmission in the CNS. Moreover, viral replication in the olfactory system and CNS is associated with inflammatory and immune responses, tissue disorganization and dysfunction. Overall, we proposed that PHEV may serve as a potential prototype for elucidating the pathogenesis of coronavirus-associated neurological complications and olfactory and taste disorders.


Assuntos
Betacoronavirus 1 , COVID-19 , Infecções por Coronavirus/patologia , Transtornos do Olfato , Animais , Betacoronavirus 1/fisiologia , Humanos , Camundongos , Transtornos do Olfato/virologia , SARS-CoV-2 , Olfato , Suínos
5.
J Virol ; 96(1): e0169521, 2022 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-34643429

RESUMO

The replication of coronaviruses, including severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), and the recently emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is closely associated with the endoplasmic reticulum (ER) of infected cells. The unfolded protein response (UPR), which is mediated by ER stress (ERS), is a typical outcome in coronavirus-infected cells and is closely associated with the characteristics of coronaviruses. However, the interaction between virus-induced ERS and coronavirus replication is poorly understood. Here, we demonstrate that infection with the betacoronavirus porcine hemagglutinating encephalomyelitis virus (PHEV) induced ERS and triggered all three branches of the UPR signaling pathway both in vitro and in vivo. In addition, ERS suppressed PHEV replication in mouse neuro-2a (N2a) cells primarily by activating the protein kinase R-like ER kinase (PERK)-eukaryotic initiation factor 2α (eIF2α) axis of the UPR. Moreover, another eIF2α phosphorylation kinase, interferon (IFN)-induced double-stranded RNA-dependent protein kinase (PKR), was also activated and acted cooperatively with PERK to decrease PHEV replication. Furthermore, we demonstrate that the PERK/PKR-eIF2α pathways negatively regulated PHEV replication by attenuating global protein translation. Phosphorylated eIF2α also promoted the formation of stress granules (SGs), which in turn repressed PHEV replication. In summary, our study presents a vital aspect of the host innate response to invading pathogens and reveals attractive host targets (e.g., PERK, PKR, and eIF2α) for antiviral drugs. IMPORTANCE Coronavirus diseases are caused by different coronaviruses of importance in humans and animals, and specific treatments are extremely limited. ERS, which can activate the UPR to modulate viral replication and the host innate response, is a frequent occurrence in coronavirus-infected cells. PHEV, a neurotropic betacoronavirus, causes nerve cell damage, which accounts for the high mortality rates in suckling piglets. However, it remains incompletely understood whether the highly developed ER in nerve cells plays an antiviral role in ERS and how ERS regulates viral proliferation. In this study, we found that PHEV infection induced ERS and activated the UPR both in vitro and in vivo and that the activated PERK/PKR-eIF2α axis inhibited PHEV replication through attenuating global protein translation and promoting SG formation. A better understanding of coronavirus-induced ERS and UPR activation may reveal the pathogenic mechanism of coronavirus and facilitate the development of new treatment strategies for these diseases.


Assuntos
Betacoronavirus 1/fisiologia , Infecções por Coronavirus/metabolismo , Fator de Iniciação 2 em Eucariotos/metabolismo , Grânulos de Estresse/metabolismo , Replicação Viral/fisiologia , eIF-2 Quinase/metabolismo , Animais , Betacoronavirus 1/metabolismo , Linhagem Celular , Infecções por Coronavirus/virologia , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/ultraestrutura , Estresse do Retículo Endoplasmático , Camundongos , Fosforilação , Biossíntese de Proteínas , Transdução de Sinais , Resposta a Proteínas não Dobradas
6.
Vet Microbiol ; 252: 108918, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33191000

RESUMO

Porcine haemagglutinating encephalomyelitis virus (PHEV) is a member of coronavirus that causes acute infectious disease and high mortality in piglets. The transcription factor IRF3 is a central regulator of type I interferon (IFN) innate immune signalling. Here, we report that PHEV infection of RAW264.7 cells results in strong suppression of IFN-ß production in the early stage. A comparative analysis of the upstream effector of IFN-ß transcription demonstrated that deactivation of IRF3, but not p65 or ATF-2 proteins, is uniquely attributed to failure of early IFN-ß induction. Moreover, the RIG-I/MDA5/MAVS/TBK1-dependent protective response that regulates the IRF3 pathway is not disrupted by PHEV and works well underlying the deactivated IRF3-mediated IFN-ß inhibition. After challenge with poly(I:C), a synthetic analogue of dsRNA used to stimulate IFN-ß secretion in the TLR-controlled pathway, we show that PHEV and poly(I:C) regulate IFN-ß-induction via two different pathways. Collectively, our findings reveal that deactivation of IRF3 is a specific mechanism that contributes to termination of type I IFN signalling during early infection with PHEV independent of the conserved RIG-I/MAVS/MDA5/TBK1-mediated innate immune response.


Assuntos
Betacoronavirus 1/imunologia , Infecções por Coronavirus/veterinária , Fator Regulador 3 de Interferon/genética , Interferon beta/imunologia , Animais , Betacoronavirus 1/genética , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Imunidade Inata , Fator Regulador 3 de Interferon/imunologia , Camundongos , Poli I-C/farmacologia , Células RAW 264.7 , Transdução de Sinais/imunologia
7.
J Gen Virol ; 96(9): 2579-2586, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26297355

RESUMO

Newcastle disease virus (NDV) causes a severe and economically significant disease affecting almost the entire poultry industry worldwide. However, factors that affect NDV replication in host cells are poorly understood. Raf kinase inhibitory protein (RKIP) is a physiological inhibitor of c-RAF kinase and NF-κB signalling, known for their functions in the control of immune response as well as tumour invasion and metastasis. In the present study, we investigated the consequences of overexpression of host RKIP during viral infection. We demonstrate that NDV infection represses RKIP expression thereby promoting virus replication. Experimental upregulation of RKIP in turn acts as a potential antiviral defence mechanism in host cells that restricts NDV replication by repressing the activation of Raf/MEK/ERK and IκBα/NF-κB signalling pathways. Our results not only extend the concept of linking NDV-host interactions, but also reveal RKIP as a new class of protein-kinase-inhibitor protein that affects NDV replication with therapeutic potential.


Assuntos
Doença de Newcastle/genética , Vírus da Doença de Newcastle/fisiologia , Proteína de Ligação a Fosfatidiletanolamina/genética , Replicação Viral , Animais , Embrião de Galinha , Regulação para Baixo , NF-kappa B/metabolismo , Doença de Newcastle/metabolismo , Doença de Newcastle/virologia , Vírus da Doença de Newcastle/genética , Proteína de Ligação a Fosfatidiletanolamina/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA