Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Immune Netw ; 22(2): e19, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35573153

RESUMO

Coxsackievirus B3 (CVB3) infection causes acute pancreatitis and myocarditis. However, its pathophysiological mechanism is unclear. Here, we investigated how lipid metabolism is associated with exacerbation of CVB3 pathology using high-fat diet (HFD)-induced obese mice. Mice were intraperitoneally inoculated with 1×106 pfu/mouse of CVB3 after being fed a control or HFD to induce obesity. Mice were treated with mitoquinone (MitoQ) to reduce the level of mitochondrial ROS (mtROS). In obese mice, lipotoxicity of white adipose tissue-induced inflammation caused increased replication of CVB3 and mortality. The coxsackievirus adenovirus receptor increased under obese conditions, facilitating CVB3 replication in vitro. However, lipid-treated cells with receptor-specific inhibitors did not reduce CVB3 replication. In addition, lipid treatment increased mitochondria-derived vesicle formation and the number of multivesicular bodies. Alternatively, we found that inhibition of lipid-induced mtROS decreased viral replication. Notably, HFD-fed mice were more susceptible to CVB3-induced mortality in association with increased levels of CVB3 replication in adipose tissue, which was ameliorated by administration of the mtROS inhibitor, MitoQ. These results suggest that mtROS inhibitors can be used as potential treatments for CVB3 infection.

2.
Immune Netw ; 20(4): e32, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32895619

RESUMO

Influenza virus is the major cause of seasonal and pandemic flu. Currently, oseltamivir, a potent and selective inhibitor of neuraminidase of influenza A and B viruses, is the drug of choice for treating patients with influenza virus infection. However, recent emergence of oseltamivir-resistant influenza viruses has limited its efficacy. Morin hydrate (3,5,7,2',4'-pentahydroxyflavone) is a flavonoid isolated from Morus alba L. It has antioxidant, anti-inflammatory, neuroprotective, and anticancer effects partly by the inhibition of the NF-кB signaling pathway. However, its effects on influenza virus have not been studied. We evaluated the antiviral activity of morin hydrate against influenza A/Puerto Rico/8/1934 (A/PR/8; H1N1) and oseltamivir-resistant A/PR/8 influenza viruses in vitro. To determine its mode of action, we carried out time course experiments, and time of addition, hemolysis inhibition, and hemagglutination assays. The effects of the co-administration of morin hydrate and oseltamivir were assessed using the murine model of A/PR/8 infection. We found that morin hydrate reduced hemagglutination by A/PR/8 in vitro. It alleviated the symptoms of A/PR/8-infection, and reduced the levels of pro-inflammatory cytokines and chemokines, such as TNF-α and CCL2, in infected mice. Co-administration of morin hydrate and oseltamivir phosphate reduced the virus titers and attenuated pulmonary inflammation. Our results suggest that morin hydrate exhibits antiviral activity by inhibiting the entry of the virus.

3.
Molecules ; 23(8)2018 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-30071639

RESUMO

Lespedeza cuneata (Fabaceae), known as Chinese bushclover, has been used in traditional medicines for the treatment of diseases including diabetes, hematuria, and insomnia. As part of a continuing search for bioactive constituents from Korean medicinal plant sources, phytochemical analysis of the aerial portion of L. cuneata led to the isolation of two new lignan glycosides (1,2) along with three known lignan glycosides (3⁻7) and nine known flavonoid glycosides (8⁻14). Numerous analysis techniques, including 1D and 2D NMR spectroscopy, CD spectroscopy, HR-MS, and chemical reactions, were utilized for structural elucidation of the new compounds (1,2). The isolated compounds were evaluated for their applicability in medicinal use using cell-based assays. Compounds 1 and 4⁻6 exhibited weak cytotoxicity against four human breast cancer cell lines (Bt549, MCF7, MDA-MB-231, and HCC70) (IC50 < 30.0 µM). However, none of the isolated compounds showed significant antiviral activity against PR8, HRV1B, or CVB3. In addition, compound 10 produced fewer lipid droplets in Oil Red O staining of mouse mesenchymal stem cells compared to the untreated negative control without altering the amount of alkaline phosphatase staining.


Assuntos
Flavonoides/química , Glicosídeos/química , Glicosídeos/farmacologia , Lespedeza/química , Lignanas/química , Animais , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacologia , Antivirais/química , Antivirais/farmacologia , Linhagem Celular Tumoral , Medicamentos de Ervas Chinesas/química , Medicamentos de Ervas Chinesas/farmacologia , Humanos , Células MCF-7 , Espectroscopia de Ressonância Magnética , Camundongos , Estrutura Molecular , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Vírus/efeitos dos fármacos
4.
J Nat Med ; 72(1): 290-297, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28914420

RESUMO

Phytochemical investigation of the methanol extract of Vitex limonifolia leaves led to the isolation of three new labdane-type diterpenoids, vitexlimolides A-C (1-3) and eight known compounds, 5,4'-dihydroxy-3,7-dimethoxyflavone (4), vitecetin (5), 5,4'-dihydroxy-7,3'-dimethoxyflavone (6), verrucosin (7), 2α, 3α-dihydroxy-urs-12-en-28-oic acid (8), euscaphlic acid (9), 18,19-seco, 2α, 3α-dihydroxy-19-oxo-urs-11,13(18)-dien-28-oic acid (10), and maslinic acid (11). Their chemical structures were elucidated by physical and chemical methods. All compounds were evaluated for antiviral activities against CVB3, HRV1B, and EV71 viruses. As a result, compounds 4 and 6 showed potent antiviral activity against CVB3 infection with IC50 values of 0.12 ± 0.06 and 1.86 ± 0.18 (µM), respectively.


Assuntos
Antivirais/isolamento & purificação , Diterpenos/isolamento & purificação , Extratos Vegetais/isolamento & purificação , Vitex/química , Animais , Antivirais/química , Antivirais/farmacologia , Chlorocebus aethiops , Diterpenos/química , Diterpenos/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Enterovirus Humano A/efeitos dos fármacos , Enterovirus Humano B/efeitos dos fármacos , Células HeLa , Humanos , Concentração Inibidora 50 , Testes de Sensibilidade Microbiana , Estrutura Molecular , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Folhas de Planta/química , Rhinovirus/efeitos dos fármacos , Triterpenos , Células Vero
5.
Antiviral Res ; 145: 6-13, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28705625

RESUMO

Rhinovirus, a major causative agent of the common cold, is associated with exacerbation of asthma and chronic obstructive pulmonary disease. Currently, there is no antiviral treatment or vaccine for human rhinovirus (HRV). Gemcitabine (2',2'-difluorodeoxycytidine, dFdC) is a deoxycytidine analog with antiviral activity against rhinovirus, as well as enterovirus 71, in vitro. However, the antiviral effects of gemcitabine in vivo have not been investigated. In the current study, we assessed whether gemcitabine mediated antiviral effects in the murine HRV infection model. Intranasal administration of gemcitabine significantly lowered pulmonary viral load and inflammation by decreasing proinflammatory cytokines, including TNF-α and IL-1ß, and reduction in the number of lung-infiltrating lymphocytes. Interestingly, we found that the addition of UTP and CTP significantly attenuated the antiviral activity of gemcitabine. Thus the limitation of UTP and CTP by the addition of gemcitabine may inhibit the viral RNA synthesis. These results suggest that gemcitabine, an antineoplastic drug, can be repositioned as an antiviral drug to inhibit HRV infection.


Assuntos
Antivirais/farmacologia , Antivirais/uso terapêutico , Desoxicitidina/análogos & derivados , Infecções por Picornaviridae/tratamento farmacológico , Rhinovirus/efeitos dos fármacos , Animais , Citocinas/biossíntese , Citocinas/imunologia , Desoxicitidina/administração & dosagem , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Modelos Animais de Doenças , Reposicionamento de Medicamentos , Humanos , Inflamação , Pulmão/imunologia , Camundongos , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/virologia , RNA Viral , Carga Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Gencitabina
6.
Oncotarget ; 8(25): 41401-41411, 2017 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-28489607

RESUMO

Trans-Scirpusin A (TSA) is a resveratrol oligomer found in Borassus flabellifer L. We found that TSA inhibited the growth of colorectal cancer Her2/CT26 cells in vivo in mice. Although some cytotoxic T lymphocytes (CTLs) were induced against the tumor-associated antigen Her2, TSA treatment did not significantly increase the level of Her2-specific CTL response compared to that with vehicle treatment. However, there was a significant increase in the level of TNF-α mRNA in tumor tissue and Her2-specific Ab (antibody) production. More importantly, we found that TSA overcomes the tumor-associated immunosuppressive microenvironment by reducing the number of CD25+FoxP3+ regulatory T cells and myeloid-derived suppressor cells (MDSCs). We detected the induction of autophagy in TSA-treated Her2/CT26 cells, based on the increased level of the mammalian autophagy protein LC3 puncta, and increased conversion of LC3-I to LC3-II. Further, TSA induced 5' AMP-activated protein kinase (p-AMPK) (T172) and inhibited mammalian target of rapamycin complex 1 (mTORC1) activity as estimated by phosphorylated ribosomal protein S6 kinase beta-1 (p-p70S6K) levels, thereby suggesting that TSA-mediated AMPK activation and inhibition of mTORC1 pathway might be associated with autophagy induction. TSA also induced apoptosis of Her2/CT26 cells, as inferred by the increased sub-G1 mitotic phases in these cells, Annexin V/PI-double positive results, and TUNEL-positive cells. Finally, we found that the combined treatment of mice with docetaxel and TSA successfully inhibited tumor growth to a greater extent than docetaxel alone. Therefore, we propose the use of TSA for supplementary anticancer therapy to support anti-neoplastic drugs, such as docetaxel, by inducing apoptosis in cancer cells and resulting in the induction of neighborhood anti-cancer immunity.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Benzofuranos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Estilbenos/farmacologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/genética , Autofagia/genética , Benzofuranos/administração & dosagem , Benzofuranos/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Docetaxel , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos Endogâmicos BALB C , Estrutura Molecular , Receptor ErbB-2/genética , Estilbenos/administração & dosagem , Estilbenos/química , Taxoides/administração & dosagem , Taxoides/farmacologia , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/genética
7.
Acta Biomater ; 53: 414-426, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28216300

RESUMO

(3-Aminomethylphenyl)boronic acid (AMPB)-installed hyaluronic acid-ceramide (HACE)-based nanoparticles (NPs), including manassantin B (MB), were fabricated for tumor-targeted delivery. The amine group of AMPB was conjugated to the carboxylic acid group of hyaluronic acid (HA) via amide bond formation, and synthesis was confirmed by spectroscopic methods. HACE-AMPB/MB NPs with a 239-nm mean diameter, narrow size distribution, negative zeta potential, and >90% drug encapsulation efficiency were fabricated. Exposed AMPB in the outer surface of HACE-AMPB NPs (in the aqueous environment) may react with sialic acid of cancer cells. The improved cellular accumulation efficiency, in vitro antitumor efficacy, and tumor penetration efficiency of HACE-AMPB/MB NPs, compared with HACE/MB NPs, in MDA-MB-231 cells (CD44 receptor-positive human breast adenocarcinoma cells) may be based on the CD44 receptor-mediated endocytosis and phenylboronic acid-sialic acid interaction. Enhanced in vivo tumor targetability, infiltration efficiency, and antitumor efficacies of HACE-AMPB NPs, compared with HACE NPs, were observed in a MDA-MB-231 tumor-xenografted mouse model. In addition to passive tumor targeting (based on an enhanced permeability and retention effect) and active tumor targeting (interaction between HA and CD44 receptor), the phenylboronic acid-sialic acid interaction can play important roles in augmented tumor targeting and penetration of HACE-AMPB NPs. STATEMENT OF SIGNIFICANCE: (3-Aminomethylphenyl)boronic acid (AMPB)-tethered hyaluronic acid-ceramide (HACE)-based nanoparticles (NPs), including manassantin B (MB), were fabricated and their tumor targeting and penetration efficiencies were assessed in MDA-MB-231 (CD44 receptor-positive human adenocarcinoma) tumor models. MB, which exhibited antitumor efficacies via the inhibition of angiogenesis and hypoxia inducible factor (HIF)-1, was entrapped in HACE-AMPB NPs in this study. Phenylboronic acid located in the outer surface of HACE-AMPB/MB NPs (in the aqueous milieu) may react with the sialic acid over-expressed in cancer cells and intramolecular B‒O bond can be formed. This phenylboronic acid-sialic acid interaction may provide additional tumor targeting and penetration potentials together with an enhanced permeability and retention (EPR) effect (passive tumor targeting) and HA-CD44 receptor interaction (active tumor targeting). Developed HACE-AMPB NP may be one of promising nanocarriers for the imaging and therapy of CD44 receptor-expressed cancers.


Assuntos
Ácidos Borônicos/química , Ácido Hialurônico/química , Nanocápsulas/química , Neoplasias Experimentais/química , Linhagem Celular Tumoral , Ceramidas/química , Reagentes de Ligações Cruzadas/química , Difusão , Humanos , Interações Hidrofóbicas e Hidrofílicas , Nanocápsulas/ultraestrutura , Nanoconjugados/química , Nanoconjugados/ultraestrutura , Neoplasias Experimentais/patologia , Tamanho da Partícula
8.
Mol Med Rep ; 14(4): 2937-44, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27573915

RESUMO

The present study investigated the photoprotective properties of the ethyl acetate fraction of Sargassum muticum (SME) against ultraviolet B (UVB)­induced skin damage and photoaging in a mouse model. HR­1 strain hairless male mice were divided into three groups: An untreated control group, a UVB­irradiated vehicle group and a UVB­irradiated SME group. The UVB­irradiated mice in the SME group were orally administered with SME (100 mg/kg body weight in 0.1 ml water per day) and then exposed to radiation at a dose of 60­120 mJ/cm2. Wrinkle formation and skin damage were evaluated by analysis of skin replicas, epidermal thickness and collagen fiber integrity in the dermal connective tissue. The mechanism underlying the action of SME was also investigated in the human HaCaT keratinocyte cell line following exposure of the cells to UVB at a dose of 30 mJ/cm2. The protein expression levels and activity of matrix metalloproteinase­1 (MMP­1), and the binding of activator protein­1 (AP­1) to the MMP­1 promoter were assessed in the HaCaT cells using western blot analysis, an MMP­1 fluorescent assay and a chromatin immune­precipitation assay, respectively. The results showed that the mean length and depth of the wrinkles in the UVB­exposed hairless mice were significantly improved by oral administration of SME, which also prevented the increase in epidermal thickness triggered by UVB irradiation. Furthermore, a marked increase in collagen bundle formation was observed in the UVB­treated mice with SME administration. SME pretreatment also significantly inhibited the UVB­induced upregulation in the expression and activity of MMP­1 in the cultured HaCaT keratinocytes, and the UVB­enhanced association of AP­1 with the MMP­1 promoter. These results suggested that SME may be useful as an anti-photoaging resource for the skin.


Assuntos
Queratinócitos/efeitos dos fármacos , Queratinócitos/efeitos da radiação , Extratos Vegetais/farmacologia , Protetores contra Radiação/farmacologia , Sargassum/química , Envelhecimento da Pele/efeitos dos fármacos , Envelhecimento da Pele/efeitos da radiação , Acetatos/química , Acetatos/farmacologia , Animais , Linhagem Celular , Humanos , Queratinócitos/citologia , Masculino , Camundongos Pelados , Extratos Vegetais/química , Protetores contra Radiação/química , Pele/citologia , Pele/efeitos dos fármacos , Pele/efeitos da radiação , Raios Ultravioleta
9.
Cancer Lett ; 381(1): 156-64, 2016 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-27431309

RESUMO

Interleukin-10 (IL-10) is a well-characterized anti-inflammatory cytokine, but its role in anti-cancer immunity is controversial. After injection with TC-1 cancer cells, we observed more rapid tumour growth and significantly higher interleukin-6 (IL-6) production in IL-10 knockout (IL-10(-/-)) mice than wild-type (WT) mice. Blocking IL-6 with an anti-IL-6 receptor (IL-6R) monoclonal antibody (mAb) inhibited tumour growth and myeloid-derived suppressor cell (MDSC) generation, which were significantly increased in IL-10-deficient mice. MDSCs and tumour cells from IL-10(-/-) mice had increased phosphorylated signal transducer and activator of transcription 3 (p-STAT3) levels. Treatment with a STAT3 inhibitor, S3I, reduced tumour growth, inhibited MDSC expansion, reduced IL-6 in tumours, and relieved T cell suppression. The combination of anti-IL-6R mAb and S3I further inhibited tumour growth compared to S3I treatment alone. These results suggested that the inhibition of the IL-6/STAT3 signalling axis is a candidate anti-cancer strategy, especially under systemic inflammatory conditions with high IL-6.


Assuntos
Proliferação de Células , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Células Supressoras Mieloides/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Neoplasias do Colo do Útero/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Genótipo , Interleucina-10/deficiência , Interleucina-10/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Supressoras Mieloides/patologia , Fenótipo , Fosforilação , Receptores de Interleucina-6/antagonistas & inibidores , Receptores de Interleucina-6/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fatores de Tempo , Carga Tumoral , Evasão Tumoral , Neoplasias do Colo do Útero/tratamento farmacológico , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/patologia
10.
Virol J ; 13: 99, 2016 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-27296985

RESUMO

BACKGROUND: Enterovirus 71 (EV71) is a major causative agent of hand-foot-mouth disease (HFMD) and also causes severe neurological complications, leading to fatality in young children. However, no effective therapy is currently available for the treatment of this infection. METHODS: We identified small-molecule inhibitors of EV71 from a screen of 968 Food and Drug Administration (FDA)-approved drugs, with which clinical application for EV71-associated diseases would be more feasible, using EV71 subgenomic replicon system. Primary hits were extensively evaluated for their antiviral activities in EV71-infected cells. RESULTS: We identified micafungin, an echinocandin antifungal drug, as a novel inhibitor of EV71. Micafungin potently inhibits the proliferation of EV71 as well as the replication of EV71 replicon in cells with a low micromolar IC50 (~5 µM). The strong antiviral effect of micafungin on EV71 replicon and the result from time-of-addition experiment demonstrated a targeting of micafungin on virion-independent intracellular process(es) during EV71 infection. Moreover, an extensive analysis excluded the involvement of 2C and 3A proteins, IRES-dependent translation, and also that of polyprotein processing in the antiviral effect of micafungin. CONCLUSIONS: Our research revealed a new indication of micafungin as an effective inhibitor of EV71, which is the first case reporting antiviral activity of micafungin, an antifungal drug.


Assuntos
Antivirais/farmacologia , Equinocandinas/farmacologia , Enterovirus Humano A/efeitos dos fármacos , Lipopeptídeos/farmacologia , Animais , Linhagem Celular , Reposicionamento de Medicamentos , Humanos , Micafungina
11.
PLoS One ; 11(5): e0155784, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27195463

RESUMO

The flavonoids mosloflavone, oroxylin A, and norwogonin, which were purified from Scutellaria baicalensis Georgi, significantly protected Vero cells against Coxsackievirus B3 (CVB3)-induced cell death. To investigate the in vivo antiviral activity of oroxylin A, we intraperitoneally inoculated CVB3 into 4-week-old BALB/c mice. Body weights and blood glucose levels of the mice were decreased after CVB3 infection, and these changes were attenuated by the administration of oroxylin A. Importantly, treatment of mice with oroxylin A reduced viral titers in the pancreas and decreased the serum levels of the inflammatory cytokines including interleukin-6 (IL-6) and tumor necrosis factor (TNF)-α. Additionally, the administration of oroxylin A mitigated the histological pancreatic lesions and apoptotic cell death induced by CVB3 infection and increased the levels of phospho-eIF2α in infected pancreata. The results suggest that oroxylin A may represent a potent antiviral agent against CVB3 infection.


Assuntos
Antivirais/farmacologia , Infecções por Coxsackievirus/tratamento farmacológico , Flavonoides/farmacologia , Pancreatopatias/tratamento farmacológico , Pancreatopatias/virologia , Animais , Apoptose , Sobrevivência Celular , Chlorocebus aethiops , Citocinas/metabolismo , Enterovirus/efeitos dos fármacos , Fator de Iniciação 2 em Eucariotos/metabolismo , Feminino , Flavonas/farmacologia , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Fosforilação , Scutellaria , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Células Vero
12.
Antiviral Res ; 124: 1-10, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26526589

RESUMO

Enteroviruses are major causative agents of various human diseases, and some of them are currently considered to be an enormous threat to public health. However, no effective therapy is currently available for the treatment of these infections. We identified gemcitabine, a nucleoside-analog drug used for cancer treatment, from a screen of bioactive chemicals as a novel inhibitor of coxsackievirus B3 (CVB3) and enterovirus 71 (EV71). Gemcitabine potently inhibited the proliferation of CVB3 and EV71, as well as the replication of CVB3 and EV71 replicons, in cells with a low micromolar IC50 (1-5 µM). Its strong inhibitory effect was also observed in cells infected with human rhinoviruses, demonstrating broad-spectrum antiviral effects on enteroviruses. Mechanistically, an extensive analysis excluded the involvement of 2C, 3A, IRES-dependent translation, and also that of polyprotein processing in the antiviral effects of gemcitabine. Importantly, gemcitabine in combination with ribavirin, an antiviral drug currently being used against a few RNA viruses, exhibited a synergistic antiviral effect on the replication of CVB3 and EV71 replicons. Consequently, our results clearly demonstrate a new indication for gemcitabine as an effective broad-spectrum inhibitor of enteroviruses and strongly suggest a new therapeutic strategy using gemcitabine alone or in combination with ribavirin for the treatment of various diseases associated with enterovirus infection.


Assuntos
Antivirais/farmacologia , Desoxicitidina/análogos & derivados , Enterovirus/efeitos dos fármacos , Ribavirina/farmacologia , Animais , Proliferação de Células , Desoxicitidina/farmacologia , Sinergismo Farmacológico , Enterovirus/fisiologia , Enterovirus Humano A/efeitos dos fármacos , Enterovirus Humano A/fisiologia , Infecções por Enterovirus/tratamento farmacológico , Infecções por Enterovirus/virologia , Células HEK293 , Células HeLa , Humanos , Replicon/efeitos dos fármacos , Células Vero , Replicação Viral/efeitos dos fármacos , Gencitabina
13.
Vaccine ; 33(48): 6604-10, 2015 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-26529069

RESUMO

Enterovirus 71 (EV71) causes hand, foot, and mouth diseases and can result in severe neurological disorders when it infects the central nervous system. Thus, there is a need for the development of effective vaccines against EV71 infection. Here we report that viral capsid protein 1 (VP1), one of the main capsid proteins of EV71, efficiently elicited VP1-specific immunoglobulin G (IgG) in the serum of mice immunized with recombinant VP1. The VP1-specific IgG produced in female mice was efficiently transferred to their offspring, conferring protection against EV71 infection immediately after birth. VP1-specific antibody can neutralize EV71 infection and protect host cells. VP1-specific maternal IgG in offspring was maintained for over 6 months. However, the pre-existence of VP1-specific maternal IgG interfered with the production of VP1-specific IgG antibody secreting cells by active immunization in offspring. Therefore, although our results showed the potential for VP1-specific maternal IgG protection against EV71 in neonatal mice, other strategies must be developed to overcome the hindrance of maternal IgG in active immunization. In this study, we developed an effective and feasible animal model to evaluate the protective efficacy of humoral immunity against EV71 infection using a maternal immunity concept.


Assuntos
Enterovirus Humano A/imunologia , Infecções por Enterovirus/prevenção & controle , Imunidade Materno-Adquirida , Imunoglobulina G/sangue , Proteínas Estruturais Virais/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Modelos Animais de Doenças , Infecções por Enterovirus/imunologia , Feminino , Imunidade Humoral , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/genética , Vacinas Virais/administração & dosagem
14.
Biomol Ther (Seoul) ; 23(4): 345-9, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26157551

RESUMO

Betulinic acid, a pentacyclic triterpene isolated from Jujube tree (Zizyphus jujuba Mill), has been known for a wide range of biological and medicinal properties such as antibacterial, antimalarial, anti-inflammatory, antihelmintic, antinociceptive, and anticancer activities. In the study, we investigated the antiviral activity on influenza A/PR/8 virus infected A549 human lung adenocarcinoma epithelial cell line and C57BL/6 mice. Betulinic acid showed the anti-influenza viral activity at a concentration of 50 µM without a significant cytotoxicity in influenza A/PR/8 virus infected A549 cells. Also, betulinic acid significantly attenuated pulmonary pathology including increased necrosis, numbers of inflammatory cells and pulmonary edema induced by influenza A/PR/8 virus infection compared with vehicle- or oseltamivir-treated mice in vivo model. The down-regulation of IFN-γ level, which is critical for innate and adaptive immunity in viral infection, after treating of betulinic acid in mouse lung. Based on the obtained results, it is suggested that betulinic acid can be the potential therapeutic agent for virus infection via anti-inflammatory activity.

15.
PLoS One ; 10(6): e0131089, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26098681

RESUMO

Several anti-influenza drugs that reduce disease manifestation exist, and although these drugs provide clinical benefits in infected patients, their efficacy is limited by the emergence of drug-resistant influenza viruses. In the current study, we assessed the therapeutic strategy of enhancing the antiviral efficacy of an existing neuraminidase inhibitor, oseltamivir, by coadministering with the leaf extract from Hedera helix L, commonly known as ivy. Ivy extract has anti-inflammatory, antibacterial, antifungal, and antihelminthic properties. In the present study, we investigated its potential antiviral properties against influenza A/PR/8 (PR8) virus in a mouse model with suboptimal oseltamivir that mimics a poor clinical response to antiviral drug treatment. Suboptimal oseltamivir resulted in insufficient protection against PR8 infection. Oral administration of ivy extract with suboptimal oseltamivir increased the antiviral activity of oseltamivir. Ivy extract and its compounds, particularly hedrasaponin F, significantly reduced the cytopathic effect in PR8-infected A549 cells in the presence of oseltamivir. Compared with oseltamivir treatment alone, coadministration of the fraction of ivy extract that contained the highest proportion of hedrasaponin F with oseltamivir decreased pulmonary inflammation in PR8-infected mice. Inflammatory cytokines and chemokines, including tumor necrosis factor-alpha and chemokine (C-C motif) ligand 2, were reduced by treatment with oseltamivir and the fraction of ivy extract. Analysis of inflammatory cell infiltration in the bronchial alveolar of PR8-infected mice revealed that CD11b+Ly6G+ and CD11b+Ly6Cint cells were recruited after virus infection; coadministration of the ivy extract fraction with oseltamivir reduced infiltration of these inflammatory cells. In a model of suboptimal oseltamivir treatment, coadministration of ivy extract fraction that includes hedrasaponin F increased protection against PR8 infection that could be explained by its antiviral and anti-inflammatory activities.


Assuntos
Antivirais/administração & dosagem , Hedera , Vírus da Influenza A/efeitos dos fármacos , Infecções por Orthomyxoviridae/tratamento farmacológico , Oseltamivir/administração & dosagem , Fitoterapia , Animais , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Citocinas/metabolismo , Efeito Citopatogênico Viral/efeitos dos fármacos , Sinergismo Farmacológico , Quimioterapia Combinada , Inibidores Enzimáticos/administração & dosagem , Técnicas In Vitro , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/patologia , Vírus da Influenza A/patogenicidade , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neuraminidase/antagonistas & inibidores , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Extratos Vegetais/administração & dosagem , Saponinas/administração & dosagem
16.
Arch Pharm Res ; 38(2): 193-202, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24752860

RESUMO

Human enterovirus 71 (EV71) and Coxsackievirus A16 (CA16) are major causative agents of hand, foot, and mouth disease (HFMD) especially in infants and children under 5 years of age. Despite recent outbreaks of HFMD, there are no approved therapeutics against EV71 and CA16 infection. Moreover, in a small percentage of cases, the disease progression can lead to serious complications of the central nervous system. In this study, we investigated the antiviral effect of corilagin and Phyllanthus urinaria extract, which contains corilagin as a major component, on EV71 and CA16 infection in vitro. Our results indicate that corilagin reduces the cytotoxicity induced by EV71 or CA16 on Vero cells with and IC50 value of 5.6 and 32.33 µg/mL, respectively. We confirmed the presence of corilagin in EtOAc and BuOH fractions from P. urinaria extract and this correlated with antiviral activity of the fractions against EV71 or CA16. Future studies will be required to confirm the antiviral activity of corilagin and P. urinaria extract in vivo. Challenging a model with a lethal dose of viral infection will be required to test this. Collectively, our work provides potential candidates for the development of novel drugs to treat HFMD.


Assuntos
Antivirais/farmacologia , Enterovirus Humano A/efeitos dos fármacos , Glucosídeos/farmacologia , Taninos Hidrolisáveis/farmacologia , Phyllanthus/química , Extratos Vegetais/farmacologia , Animais , Antivirais/isolamento & purificação , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Efeito Citopatogênico Viral/efeitos dos fármacos , Relação Dose-Resposta a Droga , Enterovirus Humano A/patogenicidade , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Células Epiteliais/virologia , Glucosídeos/isolamento & purificação , Taninos Hidrolisáveis/isolamento & purificação , Componentes Aéreos da Planta/química , Extratos Vegetais/isolamento & purificação , Células Vero
17.
Bioorg Med Chem Lett ; 24(16): 3835-40, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25066952

RESUMO

Eurycoma longifolia has been widely used for various traditional medicinal purposes in South-East Asia. In this study, five new quassinoids, eurylactone E (1), eurylactone F (2), eurylactone G (3), eurycomalide D (4), and eurycomalide E (5), along with ten known quassinoids (6-15) were isolated from the roots of E. longifolia. Their structures were determined by extensive spectroscopic methods, including 1D and 2D NMR, and MS spectra data. Among the isolated compounds, 13ß-methyl,21-dihydroeurycomanone (6) has been reported as a synthetic derivative. However, it was isolated from the natural product for the first time in this study. The cytotoxic activities of fifteen compounds were evaluated against human lung cancer cell line, A549 and human cervical cancer cell line, HeLa.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Eurycoma/química , Raízes de Plantas/química , Quassinas/farmacologia , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/isolamento & purificação , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Células HeLa , Humanos , Células MCF-7 , Estrutura Molecular , Quassinas/química , Quassinas/isolamento & purificação , Relação Estrutura-Atividade
18.
Photodermatol Photoimmunol Photomed ; 29(2): 84-9, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23458392

RESUMO

BACKGROUND: Mangifera indica L. (Anacardiaceae) is a medicinal plant whose extracts have been described as an antioxidant with anti-inflammatory and immunomodulatory activities. PURPOSE: Skin aging is a consequence of chronic sun exposure to the sun and therefore ultraviolet (UV) radiation. Naturally occurring antioxidants are known to reduce skin aging. Therefore, the aim of the present study was to evaluate the protective role of mango extract against UVB-induced skin aging in hairless mice. METHODS: HR-1 hairless male mice (6 weeks old) were divided into three groups: control (n = 5), UVB-treated vehicle (n = 5), and UVB-treated mango extract (n = 5) groups. UVB-irradiated mice from the mango extract group were orally administered 0.1 ml of water containing 100 mg of mango extract/kg body weight per day. The inhibitory activity of mango extract on wrinkle formation was determined by the analysis of the skin replica, epidermal thickness based on histological examination, and damage to collagen fiber. RESULTS: The mean length of wrinkles in UVB-treated vehicle group significantly improved after the oral administration of mango extract, which significantly inhibited the increase in epidermal thickness and epidermal hypertrophy (P < 0.05). Furthermore, a marked increase in collagen bundles was observed in the UVB-treated group after the administration of mango extract by Masson's trichrome staining. CONCLUSION: These results indicate that mango extract showed anti-photoaging activity in UVB-irradiated hairless mice.


Assuntos
Mangifera , Fitoterapia , Extratos Vegetais/farmacologia , Envelhecimento da Pele/efeitos dos fármacos , Xantonas/farmacologia , Administração Oral , Animais , Colágeno/ultraestrutura , Frutas , Masculino , Camundongos , Camundongos Pelados , Extratos Vegetais/uso terapêutico , Envelhecimento da Pele/patologia , Envelhecimento da Pele/efeitos da radiação , Raios Ultravioleta/efeitos adversos , Xantonas/uso terapêutico
19.
Virol J ; 8: 460, 2011 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-21967756

RESUMO

BACKGROUND: On the base of our previous study we were observed relevant studies on the hypothesis that the antiviral activity of quercetin 7-rhamnoside (Q7R), a flavonoid, won't relate ability of its antioxidant. METHODS: We were investigated the effects of Q7R on the cytopathic effects (CPE) by CPE reduction assay. Production of DNA fragment and reactive oxygen species (ROS) induced by PEDV infection were studied using DNA fragmentation assay and flow cytometry. RESULTS: In the course of this study it was discovered that Q7R is an extremely potent compound against PEDV. The addition of Q7R to PEDV-infected Vero cells directly reduced the formation of a visible cytopathic effect (CPE). Also, Q7R did not induce DNA fragmentation. Furthermore, ROS increased the infection of PEDV, which was strongly decreased by N-acetyl-L-cysteins (NAC). However, the increased ROS was not decreased by Q7R. Antiviral activity of antioxidants such as NAC, pyrrolidine dithiocarbamate (PDTC), and the vitamin E derivative, trolox, were hardly noticed. CONCLUSIONS: We concluded that the inhibition of PEDV production by Q7R is not simply due to a general action as an antioxidants and is highly specific, as several other antioxidants (NAC, PDTC, trolox) are inactive against PEDV infection.


Assuntos
Antivirais/farmacologia , Infecções por Coronavirus/tratamento farmacológico , Vírus da Diarreia Epidêmica Suína/efeitos dos fármacos , Quercetina/análogos & derivados , Replicação Viral/efeitos dos fármacos , Acetilcisteína/farmacologia , Animais , Antioxidantes/farmacologia , Antivirais/uso terapêutico , Chlorocebus aethiops , Cromanos/farmacologia , Infecções por Coronavirus/virologia , Efeito Citopatogênico Viral/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Citometria de Fluxo , Vírus da Diarreia Epidêmica Suína/fisiologia , Pirrolidinas/farmacologia , Quercetina/farmacologia , Quercetina/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Tiocarbamatos/farmacologia , Células Vero
20.
Phytother Res ; 24(9): 1292-6, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20104501

RESUMO

Human rhinoviruses (HRVs) are a major cause of the common cold and until now there is no registered clinically effective antiviral chemotherapeutic agent for treatment of diseases caused by HRVs. Our previous report showed that gallic acid from Woodfordia fruticosa flowers possessed antioxidant activity. Many studies reported that antioxidants possess antiviral activities against various viruses. Therefore, we examined antiviral activity of gallic acid against HRVs and mode of its actions by observing the effect of gallic acid on HRV-induced cytopathic effect (CPE) and the infectivity of HRV particles, and then carried out a time-addition study. As a result, gallic acid actively inhibited HRV2 and -3 replications with antiviral activity more than 55% without cytotoxicity in human epitheloid carcinoma cervix (HeLa) cells at a concentration of 100 mug/mL. Also, ribavirin showed lower anti-HRV3 activity than gallic acid and similar anti-HRV3 activity to it. The addition of gallic acid to HRV-infected HeLa cells directly reduced the formation of a visible CPE. Furthermore, gallic acid did directly interact or activate with HRV particles. Collectively, we concluded that the inhibition of HRV production by gallic acid is mainly due to a general action as an antioxidant and the mode of action derived from the inhibition of virus absorption.


Assuntos
Antioxidantes/uso terapêutico , Antivirais/uso terapêutico , Ácido Gálico/uso terapêutico , Infecções por Picornaviridae/tratamento farmacológico , Extratos Vegetais/uso terapêutico , Rhinovirus/efeitos dos fármacos , Woodfordia/química , Antioxidantes/farmacologia , Antivirais/farmacologia , Linhagem Celular Tumoral , Flores , Ácido Gálico/farmacologia , Células HeLa , Humanos , Fitoterapia , Infecções por Picornaviridae/virologia , Extratos Vegetais/farmacologia , Rhinovirus/patogenicidade , Ribavirina/farmacologia , Ribavirina/uso terapêutico , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA