Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Environ Sci (China) ; 147: 359-369, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39003053

RESUMEN

Agricultural practices significantly contribute to greenhouse gas (GHG) emissions, necessitating cleaner production technologies to reduce environmental pressure and achieve sustainable maize production. Plastic film mulching is commonly used in the Loess Plateau region. Incorporating slow-release fertilizers as a replacement for urea within this practice can reduce nitrogen losses and enhance crop productivity. Combining these techniques represents a novel agricultural approach in semi-arid areas. However, the impact of this integration on soil carbon storage (SOCS), carbon footprint (CF), and economic benefits has received limited research attention. Therefore, we conducted an eight-year study (2015-2022) in the semi-arid northwestern region to quantify the effects of four treatments [urea supplied without plastic film mulching (CK-U), slow-release fertilizer supplied without plastic film mulching (CK-S), urea supplied with plastic film mulching (PM-U), and slow-release fertilizer supplied with plastic film mulching (PM-S)] on soil fertility, economic and environmental benefits. The results revealed that nitrogen fertilizer was the primary contributor to total GHG emissions (≥71.97%). Compared to other treatments, PM-S increased average grain yield by 12.01%-37.89%, water use efficiency by 9.19%-23.33%, nitrogen accumulation by 27.07%-66.19%, and net return by 6.21%-29.57%. Furthermore, PM-S decreased CF by 12.87%-44.31% and CF per net return by 14.25%-41.16%. After eight years, PM-S increased SOCS (0-40 cm) by 2.46%, while PM-U decreased it by 7.09%. These findings highlight the positive effects of PM-S on surface soil fertility, economic gains, and environmental benefits in spring maize production on the Loess Plateau, underscoring its potential for widespread adoption and application.


Asunto(s)
Agricultura , Huella de Carbono , Fertilizantes , Plásticos , Zea mays , Zea mays/crecimiento & desarrollo , Agricultura/métodos , China , Suelo/química , Gases de Efecto Invernadero/análisis , Nitrógeno/análisis
2.
MedComm (2020) ; 5(4): e531, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38617435

RESUMEN

Pyrogallol, a natural polyphenol compound (1,2,3-trihydroxybenzene), has shown efficacy in the therapeutic treatment of disorders associated with inflammation. Nevertheless, the mechanisms underlying the protective properties of pyrogallol against influenza A virus infection are not yet established. We established in this study that pyrogallol effectively alleviated H1N1 influenza A virus-induced lung injury and reduced mortality. Treatment with pyrogallol was found to promote the expression and nuclear translocation of nuclear factor erythroid-2-related factor 2 (Nrf2) and peroxisome proliferator-activated receptor gamma (PPAR-γ). Notably, the activation of Nrf2 by pyrogallol was involved in elevating the expression of PPAR-γ, both of which act synergistically to enhance heme oxygenase-1 (HO-1) synthesis. Blocking HO-1 by zinc protoporphyrin (ZnPP) reduced the suppressive impact of pyrogallol on H1N1 virus-mediated aberrant retinoic acid-inducible gene-I-nuclear factor kappa B (RIG-I-NF-κB) signaling, which thus abolished the dampening effects of pyrogallol on excessive proinflammatory mediators and cell death (including apoptosis, necrosis, and ferroptosis). Furthermore, the HO-1-independent inactivation of janus kinase 1/signal transducers and activators of transcription (JAK1/STATs) and the HO-1-dependent RIG-I-augmented STAT1/2 activation were both abrogated by pyrogallol, resulting in suppression of the enhanced transcriptional activity of interferon-stimulated gene factor 3 (ISGF3) complexes, thus prominently inhibiting the amplification of the H1N1 virus-induced proinflammatory reaction and apoptosis in interferon-beta (IFN-ß)-sensitized cells. The study provides evidence that pyrogallol alleviates excessive proinflammatory responses and abnormal cell death via HO-1 induction, suggesting it could be a potential agent for treating influenza.

3.
Chin Med ; 18(1): 139, 2023 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-37891648

RESUMEN

BACKGROUND: Rosmarinic acid (RosA) is a natural phenolic compound that possesses a wide-range of pharmacological properties. However, the effects of RosA on influenza A virus-mediated acute lung injury remain unknown. In this study, we aimed to explore whether RosA could protect against H1N1 virus-mediated lung injury and elucidate the underlying mechanisms. METHODS: Mice were intragastrically administered with RosA for 2 days before intranasal inoculation of the H1N1 virus (5LD50) for the establishment of an acute lung injury model. At day 7 post-infection (p.i.), gross anatomic lung pathology, lung histopathologic, and lung index (lung weight/body weight) were examined. Luminex assay, multiple immunofluorescence and flow cytometry were performed to detect the levels of pro-inflammatory cytokines and apoptosis, respectively. Western blotting and plasmid transfection with hematopoietic-type PGD2 synthase (h-PGDS) overexpression were conducted to elucidate the mechanisms. RESULTS: RosA effectively attenuated H1N1 virus-triggered deterioration of gross anatomical morphology, worsened lung histopathology, and elevated lung index. Excessive pro-inflammatory reactions, aberrant alveolar epithelial cell apoptosis, and cytotoxic CD8+ T lung recruitment in the lung tissues induced by H1N1 virus infection were observed to be reduced by RosA treatment. In vitro experiments demonstrated that RosA treatment dose-dependently suppressed the increased levels of pro-inflammatory mediators and apoptosis through inhibition of nuclear factor kappa B (NF-κB) and P38 MAPK signaling pathways in H1N1 virus-infected A549 cells, which was accompanied by promoting activation of the h-PGDS-PGD2-HO-1 signal axis. Furthermore, we strikingly found that h-PGDS inhibition significantly abrogated the inhibitory effects of RosA on H1N1 virus-mediated activation of NF-κB and P38 MAPK signaling pathways, resulting in diminishing the suppressive effects on the increased levels of pro-inflammatory cytokines and chemokines as well as apoptosis. Finally, suppressing h-PGDS prominently abolished the protective effects of RosA on H1N1 virus-mediated severe pneumonia and lung injury. CONCLUSIONS: Taken together, our study demonstrates that RosA is a promising compound to alleviate H1N1 virus-induced severe lung injury through prompting the h-PGDS-PGD2-HO-1 signal axis.

4.
Food Funct ; 14(7): 3357-3378, 2023 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-36942763

RESUMEN

The severity of a viral respiratory illness was greatly exacerbated after exposure to a contaminant containing benzo[a]pyrene (B[a]P). Flavonoid-rich fruit intake has gained intense interest due to their health-promoting benefits for viral respiratory diseases, including influenza viruses. In our study, diosmetin (3',5,7-trihydroxy-4'-methoxyflavone), a naturally occurring hydroxylated methoxyflavone that is abundant in Citrus fruits, was explored for its effects on B[a]P-exacerbated H1N1 influenza virus-mediated inflammation and lung injury. Initially, in vivo results demonstrated that diosmetin protected against H1N1 virus-elicited acute lung injury. Simultaneously, H1N1 virus or B[a]P-stimulated A549 cells treated with diosmetin inhibited NF-κB and P-P38 activation, resulting in suppression of pro-inflammatory cytokines and apoptosis. Interestingly, diosmetin obviously promoted the expression of PPAR-γ as well as nuclear translocation of PPAR-γ, whereas, PPAR-γ inhibition by GW9662 weakened the inhibitory effects of diosmetin on H1N1 virus or B[a]P-mediated activation of NF-κB and P-P38, elevated expression of pro-inflammatory mediators as well as apoptosis. Furthermore, it was surprising to discover that mice exposed to both B[a]P and H1N1 viruses contributed to exacerbated acute lung injury, which were significantly ameliorated by diosmetin administration. In vitro studies showed that A549 cells with the combination of B[a]P and H1N1 virus augmented NF-κB and P-P38 activation, accompanied by higher levels of pro-inflammatory mediators and apoptosis, all of which were also significantly reduced by diosmetin treatment. Repressing PPAR-γ abrogated the inhibitory effects of diosmetin on B[a]P-exacerbated H1N1 virus-mediated NF-κB and P-P38 activation, inflammation, and apoptosis in A549 cells. Our findings suggest that diosmetin protected against B[a]P-exacerbated H1N1 virus-mediated lung injury by suppressing the exacerbation of NF-κB and P38 kinase activation in a PPAR-γ-dependent manner, suggesting potential benefits for B[a]P-exacerbated influenza-related illness therapeutics.


Asunto(s)
Lesión Pulmonar Aguda , Subtipo H1N1 del Virus de la Influenza A , Gripe Humana , Animales , Ratones , Humanos , FN-kappa B/metabolismo , Benzo(a)pireno , PPAR gamma/genética , PPAR gamma/metabolismo , Inflamación/tratamiento farmacológico , Flavonoides/farmacología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/tratamiento farmacológico , Gripe Humana/tratamiento farmacológico , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Mediadores de Inflamación
5.
J Inflamm (Lond) ; 19(1): 24, 2022 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-36451220

RESUMEN

BACKGROUND: Acute lung injury (ALI) arises from sepsis or bacterial infection, which are life-threatening respiratory disorders that cause the leading cause of death worldwide. 5-Methoxyflavone, a methylated flavonoid, is gaining increased attention for its various health benefits. In the current study, we investigated the potential effects of 5-methoxyflavone against LPS-mediated ALI and elucidated the corresponding possible mechanism. METHODS: A mouse model with ALI was established by intratracheal instillation of LPS, and lung pathological changes, signaling pathway related proteins and apoptosis in lung tissues were estimated by H&E staining, immunofluorescence and TUNEL assay, respectively. Cell viability was evaluated by MTT assay; protein levels of pro-inflammatory mediators were measured by ELISA assay; levels of ROS and M1 macrophage polarization were assayed by flow cytometry; the expression of Nrf2 signaling, NOX4/TLR4 axis and P-STAT1 were detected by western blotting. RESULTS: Our results showed that 5-methoxyflavone treatment inhibited LPS-induced expression of NOX4 and TLR4 as well as the activation of downstream signaling (NF-κB and P38 MAPK), which was accompanied by markedly decreased ROS levels and pro-inflammatory cytokines (IL-6, TNF-α, MCP-1, and IL-8) in BEAS-2B cells. Moreover, we revealed that these effects of 5-methoxyflavone were related to its Nrf2 activating property, and blockade of Nrf2 prevented its inhibitory effects on NOX4/TLR4/NF-κB/P38 MAPK signaling, thus abrogating the anti-inflammatory effects of 5-methoxyflavone. Besides, the Nrf2 activating property of 5-methoxyflavone in RAW264.7 cells led to inhibition of LPS/IFN-γ-mediated STAT1 signaling, resulting in suppression of LPS/IFN-γ-induced M1 macrophage polarization and the repolarization of M2 macrophages to M1. In a mouse model of LPS-induced ALI, 5-methoxyflavone administration ameliorated LPS-mediated lung pathological changes, the increased lung index (lung/body weight ratio), and epithelial cell apoptosis. Meanwhile, we found 5-methoxyflavone effectively suppressed the hyperactive signaling pathways and the production of excessive pro-inflammatory mediators. Moreover, 5-methoxyflavone reduced LPS-mediated M1 macrophage polarization associated with elevated P-STAT1 activation in the lung tissues. In addition, 5-methoxyflavone improved the survival of LPS-challenged mice. CONCLUSION: These results indicated that 5-methoxyflavone might be suitable for the development of a novel drug for ALI therapeutic.

6.
Cell Mol Biol Lett ; 27(1): 82, 2022 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-36180831

RESUMEN

Influenza-related acute lung injury (ALI) is a life-threatening condition that results mostly from uncontrolled replication of influenza virus (IV) and severe proinflammatory responses. The methoxy flavonoid compound 5-methoxyflavone (5-MF) is believed to have superior biological activity in the treatment of cancer. However, the effects and underlying mechanism of 5-MF on IV-mediated ALI are still unclear. Here, we showed that 5-MF significantly improved the survival of mice with lethal IV infection and ameliorated IV-mediated lung edema, lung histological changes, and inflammatory cell lung recruitment. We found that 5-MF has antiviral activity against influenza A virus (IAV), which was probably associated with increased expression of radical S-adenosyl methionine domain containing 2 (RSAD2) and suppression of endosomal acidification. Moreover, IV-infected A549 cells with 5-MF treatment markedly reduced proinflammatory mediator expression (IL-6, CXCL8, TNF-α, CXCL10, CCL2, CCL3, CCL4, GM-CSF, COX-2, and PGE2) and prevented P-IKBα, P-P65, and P-P38 activation. Interestingly, we demonstrated that 5-MF treatment could trigger activation of AMP-activated protein kinase (AMPK)α in IV-infected A549 cells, as evidenced by activation of the AMPKα downstream molecule P53. Importantly, the addition of AMPKα blocker compound C dramatically abolished 5-MF-mediated increased levels of RSAD2, the inhibitory effects on H1N1 virus-elicited endosomal acidification, and the suppression expression of proinflammatory mediators (IL-6, TNF-α, CXCL10, COX-2 and PGE2), as well as the inactivation of P-IKBα, P-P65, and P-P38 MAPK signaling pathways. Furthermore, inhibition of AMPKα abrogated the protective effects of 5-MF on H1N1 virus-mediated lung injury and excessive inflammation in vivo. Taken together, these results indicate that 5-MF alleviated IV-mediated ALI and suppressed excessive inflammatory responses through activation of AMPKα signaling.


Asunto(s)
Lesión Pulmonar Aguda , Subtipo H1N1 del Virus de la Influenza A , Virus de la Influenza A , Proteínas Quinasas Activadas por AMP/metabolismo , Lesión Pulmonar Aguda/metabolismo , Animales , Antivirales/farmacología , Ciclooxigenasa 2 , Flavonas , Flavonoides/farmacología , Flavonoides/uso terapéutico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Inflamación/tratamiento farmacológico , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Virus de la Influenza A/metabolismo , Interleucina-6/metabolismo , Metionina/farmacología , Metionina/uso terapéutico , Ratones , FN-kappa B/metabolismo , Prostaglandinas E/farmacología , Prostaglandinas E/uso terapéutico , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
BMC Complement Med Ther ; 21(1): 289, 2021 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-34836523

RESUMEN

BACKGROUND: H9N2 avian influenza viruses (AIVs) infect avian and mammalian hosts and provide internal genes for new emerging highly pathogenic avian viruses that cause severe pneumonia with high mortality, for which few medications are available. Arctiin, a bioactive lignan glycoside, has been reported to possess multiple pharmacological properties. However, the effect of arctiin on H9N2 virus infection is unclear. In the current study, we analyzed the effect of arctiin on H9N2 virus infection and the underlying molecular mechanism in vitro. METHODS: The antiviral effect against H9N2 virus was determined by plaque reduction assay (PRA) and progeny virus reduction assay. We employed MTT assay, qRT-PCR, ELISA, immunofluorescence and Western blotting to better understand the anti-inflammatory effect and corresponding mechanism of arctiin on H9N2 virus-infected cells. RESULTS: The results showed that arctiin had antiviral activity against H9N2 virus. Arctiin treatment reduced H9N2 virus-triggered proinflammatory cytokines, such as IL-6, and TNF-α. Moreover, arctiin significantly suppressed H9N2 virus-mediated expression of COX-2 and PGE2. Furthermore, we found that arctiin inhibited H9N2 virus-mediated activation of RIG-I/JNK MAPK signaling. Interestingly, arctiin treatment obviously reversed H9N2 virus-induced reduction of Nrf2, increased the nuclear translocation of Nrf2, and upregulated Nrf2 signaling target genes (HO-1 and SOD2). Zinc protoporphyrin (Znpp)-an HO-1 inhibitor-weakened the inhibitory effect of arctiin on H9N2 virus-induced RIG-I/JNK MAPK and proinflammatory mediators. CONCLUSION: Taken together, these results suggested that the anti-inflammatory effects of arctiin on H9N2 virus infection may be due to the activation of Nrf2/HO-1 and blocked RIG-I/JNK MAPK signaling; thus, arctiin may be a promising agent for prevention and treatment of H9N2 virus infections.


Asunto(s)
Antiinflamatorios/farmacología , Furanos/farmacología , Glucósidos/farmacología , Hemo-Oxigenasa 1/metabolismo , Subtipo H9N2 del Virus de la Influenza A/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Extractos Vegetales/farmacología , Células A549 , Animales , Antiinflamatorios/química , China , Perros , Furanos/química , Glucósidos/química , Humanos , Células de Riñón Canino Madin Darby , Estructura Molecular
8.
Acta Pharmacol Sin ; 41(9): 1178-1196, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32504068

RESUMEN

ß-Sitosterol (24-ethyl-5-cholestene-3-ol) is a common phytosterol Chinese medical plants that has been shown to possess antioxidant and anti-inflammatory activity. In this study we investigated the effects of ß-sitosterol on influenza virus-induced inflammation and acute lung injury and the molecular mechanisms. We demonstrate that ß-sitosterol (150-450 µg/mL) dose-dependently suppresses inflammatory response through NF-κB and p38 mitogen-activated protein kinase (MAPK) signaling in influenza A virus (IAV)-infected cells, which was accompanied by decreased induction of interferons (IFNs) (including Type I and III IFN). Furthermore, we revealed that the anti-inflammatory effect of ß-sitosterol resulted from its inhibitory effect on retinoic acid-inducible gene I (RIG-I) signaling, led to decreased STAT1 signaling, thus affecting the transcriptional activity of ISGF3 (interferon-stimulated gene factor 3) complexes and resulting in abrogation of the IAV-induced proinflammatory amplification effect in IFN-sensitized cells. Moreover, ß-sitosterol treatment attenuated RIG-I-mediated apoptotic injury of alveolar epithelial cells (AEC) via downregulation of pro-apoptotic factors. In a mouse model of influenza, pre-administration of ß-sitosterol (50, 200 mg·kg-1·d-1, i.g., for 2 days) dose-dependently ameliorated IAV-mediated recruitment of pathogenic cytotoxic T cells and immune dysregulation. In addition, pre-administration of ß-sitosterol protected mice from lethal IAV infection. Our data suggest that ß-sitosterol blocks the immune response mediated by RIG-I signaling and deleterious IFN production, providing a potential benefit for the treatment of influenza.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Antivirales/uso terapéutico , Proteína 58 DEAD Box/metabolismo , Inflamación/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Sitoesteroles/uso terapéutico , Células A549 , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/virología , Animales , Antivirales/análisis , Apoptosis/efectos de los fármacos , Perros , Femenino , Células HEK293 , Humanos , Inflamación/patología , Inflamación/virología , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Interferón Tipo I/metabolismo , Interferones/metabolismo , Pulmón/patología , Células de Riñón Canino Madin Darby , Ratones Endogámicos BALB C , Plantas/química , Factor de Transcripción STAT1/metabolismo , Sitoesteroles/análisis , Interferón lambda
9.
J Pharm Anal ; 10(2): 130-146, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32373385

RESUMEN

Isatis indigotica Fort. (Ban-Lan-Gen) is an herbal medicine prescribed for influenza treatment. However, its active components and mode of action remain mostly unknown. In the present study, erucic acid was isolated from Isatis indigotica Fort., and subsequently its underlying mechanism against influenza A virus (IAV) infection was investigated in vitro and in vivo. Our results demonstrated that erucic acid exhibited broad-spectrum antiviral activity against IAV resulting from reduction of viral polymerase transcription activity. Erucic acid was found to exert inhibitory effects on IAV or viral (v) RNA-induced pro-inflammatory mediators as well as interferons (IFNs). The molecular mechanism by which erucic acid with antiviral and anti-inflammatory properties was attributed to inactivation of NF-κB and p38 MAPK signaling. Furthermore, the NF-κB and p38 MAPK inhibitory effect of erucic acid led to diminishing the transcriptional activity of interferon-stimulated gene factor 3 (ISGF-3), and thereby reducing IAV-triggered pro-inflammatory response amplification in IFN-ß-sensitized cells. Additionally, IAV- or vRNA-triggered apoptosis of alveolar epithelial A549 cells was prevented by erucic acid. In vivo, erucic acid administration consistently displayed decreased lung viral load and viral antigens expression. Meanwhile, erucic acid markedly reduced CD8+ cytotoxic T lymphocyte (CTL) recruitment, pro-apoptotic signaling, hyperactivity of multiple signaling pathways, and exacerbated immune inflammation in the lung, which resulted in decreased lung injury and mortality in mice with a mouse-adapted A/FM/1/47-MA(H1N1) strain infection. Our findings provided a mechanistic basis for the action of erucic acid against IAV-mediated inflammation and injury, suggesting that erucic acid may have a therapeutic potential in the treatment of influenza.

10.
BMC Complement Med Ther ; 20(1): 135, 2020 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-32370749

RESUMEN

BACKGROUND: Human respiratory system infected with influenza A virus (IAV) elicited a robust pro-inflammatory response that resulted in severe illness and even death. Currently, limited immunomodulator is available to counteract IAV-associated pneumonia in the clinic. Sinensetin, a polymethoxylated flavone with five methoxy groups, has been found to possess anti-agiogenesis, anti-inflammatory and anti-diabetic activities. However, the effects of sinensetin on IAV-triggered pro-inflammatory response remain unclear. In the present study, the anti-inflammatory effects and corresponding possible mechanism of sinensetin in IAV-infected A549 cells were subjected to investigations. METHODS: The cytotoxic effects of sinensetin towards A549 cells was detected by MTT and LDH assays. The antiviral activity of sinensetin against influenza A virus was assayed in A549 cells with an engineered replication-competent influenza A virus carrying Gaussia luciferase reporter gene infection. The effect of sinensetin on influenza A virus-triggered inflammatory reaction was determined by qRT-PCR, Luminex assays, ELISA and Western blot. RESULTS: Our results showed that sinensetin did not exhibit antiviral activity against A/PR/8/34 (H1N1). Meanwhile, sinensetin treatment significantly decreased IAV-induced expression of pro-inflammatory mediators at mRNA and protein levels, including IL-6, TNF-α, IP-10, IL-8 and MCP-1. Additionally, levels of cyclooxygenase (COX)-2 and the downstream product prostaglandin E2 (PGE2) up-regulated by IAV infection were dramatically suppressed by sinensetin. The mechanistic investigation revealed that sinensetin treatment suppressed the NF-κB transcriptional activity using the NF-κB reporter stable HEK293 cell line stimulated with TNF-α (20 ng/mL) or influenza H1N1 virus. Furthermore, sinensetin abrogated influenza H1N1 virus-induced activation of NF-κB, ERK1/2 MAPK and p38 MAPK signalings. CONCLUSION: Collectively, our results indicated that sinensetin has potential capacity to attenuate IAV-triggered pro-inflammatory response via inactivation of NF-κB, ERK1/2 MAPK and p38 MAPK signalings, which implied that sinensetin may be a promising candidate drug for influenza H1N1 virus infection therapeutics.


Asunto(s)
Antivirales/farmacología , Flavonoides/farmacología , Inflamación/virología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , FN-kappa B/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Células A549 , Animales , Antivirales/química , Perros , Flavonoides/química , Humanos , Subtipo H1N1 del Virus de la Influenza A , Células de Riñón Canino Madin Darby
11.
Eur J Pharmacol ; 860: 172543, 2019 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-31323223

RESUMEN

Ergosterol peroxide has been shown to exhibit anti-tumor, antioxidant and anti-bacterial properties. However, the effects of ergosterol peroxide isolated from the herbal Baphicacanthus cusia root on influenza virus infection remain poorly understood. In the present study, ergosterol peroxide (compound 22) was obtained from the B. cusia root and subjected to investigation regarding its immunoregulatory effect on influenza A virus (IAV)-induced inflammation in A549 human alveolar epithelial cells. The structure of compound 22 isolated from B. cusia root. was elucidated by NMR analyses. Structure determination showed that the chemical structure of compound 22 closely resembles that of ergosterol peroxide. We observed that ergosterol peroxide treatment significantly suppressed IAV-induced upregulation of RIG-I expression. Additionally, ergosterol peroxide inhibited the activation of RIG-I downstream signaling pathways, including p38 MAP kinase and NF-κB, which ultimately resulted in the reduced production of an array of pro-inflammatory mediators and interferons (IFN-ß and IFN-λ1). Interestingly, inhibitory effects of ergosterol peroxide on the expression of IFNs did not affect the expression of antiviral effectors or enhance viral replication. On the other hand, ergosterol peroxide effectively abolished the amplified production of pro-inflammatory mediators in cells pretreated with IFN-ß (500 ng/ml) prior to IAV infection. Moreover, Annexin V and Hoechst 33258 staining revealed that increased apoptosis of IAV-infected cells was reversed by the presence of ergosterol peroxide. Our findings suggest that ergosterol peroxide from the B. cusia root suppressed IAV-associated inflammation and apoptosis via blocking RIG-I signaling, which may serve as a supplementary approach to the treatment of influenza.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteína 58 DEAD Box/metabolismo , Ergosterol/análogos & derivados , Subtipo H1N1 del Virus de la Influenza A/fisiología , Transducción de Señal/efectos de los fármacos , Células A549 , Animales , Perros , Ergosterol/química , Ergosterol/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/patología , Inflamación/virología , Mediadores de Inflamación/metabolismo , Interferones/biosíntesis , Células de Riñón Canino Madin Darby , Receptores Inmunológicos
12.
Mol Med Rep ; 19(1): 563-572, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30483751

RESUMEN

Eucommia ulmoides Oliver (Du-Zhong) is an ancient Chinese herbal remedy used for the treatment of various diseases. To date, the effects of its constituent lignans on influenza viruses remain to be elucidated. In the present study, a lignan glycoside was isolated and purified from Eucommia ulmoides Oliver. Its structures were identified via extensive spectroscopic analysis, and its antiviral and anti­inflammatory activities, specifically against influenza viruses, were determined via a cytopathic effect (CPE) assay, plaque­reduction assays, a progeny virus yield reduction assay, reverse transcription­quantitative polymerase chain reaction analysis and a Luminex assay. Additionally, western blot analysis was performed to investigate the underlying mechanisms of its effects against influenza viruses. The chemical and spectroscopic methods determined the structure of lignan glycoside to be (+)­pinoresinol­O­ß­D­glucopyranoside. The CPE assay showed that (+)­pinoresinol­O­ß­D­glucopyranoside exerted inhibitory activities with 50% inhibition concentration values of 408.81±5.24 and 176.24±4.41 µg/ml against the influenza A/PR/8/34 (H1N1) and A/Guangzhou/GIRD07/09 (H1N1) strains, respectively. Its antiviral properties were confirmed by plaque reduction and progeny virus yield reduction assays. Additional mechanistic analyses indicated that the anti­H1N1 virus­induced effects of (+)­pinoresinol­O-ß­D-glucopyranoside were likely due to inactivation of the nuclear factor­κB, p38 mitogen­activated protein kinase and AKT signaling pathways. Furthermore, (+)­pinoresinol­O­ß­D­glucopyranoside exhibited pronounced inhibitory effects on the expression of influenza H1N1 virus­induced pro­inflammatory mediators, including tumor necrosis factor­α, interleukin (IL)­6, IL­8 and monocyte chemoattractant protein 1. The data obtained suggest that (+)­pinoresinol­O­ß­D-glucopyranoside may be a candidate drug for treating influenza H1N1 virus infection.


Asunto(s)
Antiinflamatorios/farmacología , Antivirales/farmacología , Eucommiaceae/química , Furanos/farmacología , Glicósidos/farmacología , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Gripe Humana/tratamiento farmacológico , Lignanos/farmacología , Células A549 , Animales , Línea Celular , Línea Celular Tumoral , Perros , Humanos , Células de Riñón Canino Madin Darby , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Extractos Vegetales/farmacología , Transducción de Señal/efectos de los fármacos
13.
Mol Med Rep ; 18(1): 523-531, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29749442

RESUMEN

The influenza virus is a pathogen that can cause pandemic and epidemic outbreaks, and therefore represents a severe threat to human health. Antiviral drugs have an important role in the prevention and treatment of influenza, although the increasing emergence of drug resistance has given rise to a requirement for the development of novel antiviral drugs. In the present study, an active component (C8) isolated from Laggera pterodonta was evaluated. The nuclear magnetic resonance spectroscopy and mass spectrometry analysis results revealed that two eudesmane­type sesquiterpene compounds were identified in C8; pterodontic acid and pterodondiol. C8 was demonstrated to have a broad­spectrum effect against different influenza viruses, including human and avian influenza viruses, with a half maximal inhibitory concentration value of 19.9­91.4 µg/ml. The antiviral mechanisms of C8 were further clarified. Western blot analysis verified that C8 inhibited Toll­like receptor 7, myeloid differentiation primary response protein 88 and tumor necrosis factor (TNF) receptor associated factor 6 expression, in addition to p65 phosphorylation, at a concentration of 100 or 150 µg/ml. An indirect immunofluorescence assay demonstrated that C8 may inhibit p65/NF­κB nuclear translocation. Additionally, C8 prevented an increase in cytokine mRNA expression, including interleukin (IL)­1ß, IL­6, IL­8 and C­C motif chemokine 2 (MCP­1). Furthermore, the Bio­Plex assay results indicated that the protein expression of IL­6, IL­8, TNF­α, C­X­C motif chemokine 10, MCP­1 and C­C motif chemokine 5 was inhibited. These findings suggest that C8 has the potential to be developed into an anti­inflammatory drug for the prevention and treatment of influenza A virus infection.


Asunto(s)
Antivirales/farmacología , Células Epiteliales/virología , Virus de la Influenza A/efectos de los fármacos , Sesquiterpenos/farmacología , Transducción de Señal/efectos de los fármacos , Células A549 , Animales , Antivirales/uso terapéutico , Asteraceae/química , Línea Celular , Perros , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Sesquiterpenos/aislamiento & purificación , Sesquiterpenos/uso terapéutico , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo
14.
Molecules ; 22(10)2017 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-29035328

RESUMEN

Laggera pterodonta (DC.) Benth. is a traditional Chinese medicine. The previous study revealed that the crude extracts of this herb could inhibit influenza virus infection, but its anti-influenza components and underlying mechanism of action remain unknown. Column chromatography was performed to isolate components from the plant. Activity against influenza virus of the compound was determined by CPE inhibition assay. Neuraminidase (NA) inhibition was measured by chemiluminescence assay. The anti-virus and anti-inflammation effects were determined using dual-luciferase reporter assay, immunofluorescence, quantitative real-time PCR and luminex assay. Pterodontic acid was isolated from L. pterodonta, which showed selective anti-viral activities to H1 subtype of human influenza A virus. Meanwhile, the NA activity was not obviously inhibited by the compound. Further experiments exhibited that the compound can suppress the activation of NF-κB signal pathway and export of viral RNP complexes from the nucleus. In addition, it can significantly attenuate expression of the pro-inflammatory molecules IL-6, MIP-1ß, MCP-1, and IP-10 induced by human influenza A virus (H1N1) and similarly downregulate expression of cytokines and chemokines induced by avian influenza A virus (H9N2). This study showed that in vitro antiviral activity of pterodontic acid is most probably associated with inhibiting the replication of influenza A virus by blocking nuclear export of viral RNP complexes, and attenuating the inflammatory response by inhibiting activation of the NF-κB pathway. Pterodontic acid might be a potential antiviral agent against influenza A virus.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Asteraceae/química , Sesquiterpenos/química , Sesquiterpenos/farmacología , Línea Celular , Quimiocina CCL2/metabolismo , Quimiocina CCL4/metabolismo , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/farmacología , Células HEK293 , Humanos , Inflamación/metabolismo , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H9N2 del Virus de la Influenza A/efectos de los fármacos , Interleucina-6/metabolismo , Replicación Viral/efectos de los fármacos
15.
Acta Pharmacol Sin ; 28(11): 1717-23, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17959021

RESUMEN

AIM: This study was to investigate the effect of dehydroevodiamine (DHED) on Alzheimer's disease (AD)-like tau hyperphosphorylation induced by calyculin A (CA), an inhibitor of protein phosphatase (PP)-2A and PP-1, and the involvement of PP-2A in metabolically competent rat brain slices. METHODS: Rat brain slices were pre-incubated at 33 degree centigrade in the presence (10, 100, and 200 micromol/L, respectively) or absence of DHED for 1 h. Then, CA 0.1 micromol/L was added and the slices were treated for another 2 h. Western blotting and/or immunohistochemistry were used to measure the phosphorylation level of tau and PP-2A. RESULTS: CA treatment could remarkably increase the immunoreactivity of pS262 and decrease the staining of Tau-1, representing tau hyperphosphorylation at Ser262 (pS262) and Ser198/ 199/202 (Tau-1, as the antibody reacts with unphosphorylated tau, therefore, decreased staining represents increased phosphorylation). Pre-incubation of the brain slices with DHED could efficiently attenuate the CA-induced tau hyperphosphorylation at the above AD-related sites. Additionally, DHED also decreased the basal phosphorylation level of tau at Ser396, although CA failed to induce tau hyperphosphorylation at this site. Furthermore, CA treatment induced an increased level of Tyr307-phosphorylated PP-2A, which represents inactivation of the phosphatase, whereas DHED arrested the elevation of the inhibitory modification of PP-2A. CONCLUSION: DHED can attenuate CA-induced tau hyperphosphorylation at multiple AD-related sites in metabolically active rat brain slices. The underlying mechanism may involve a decreased inhibitory phosphorylation of PP-2A at Tyr307.


Asunto(s)
Alcaloides/farmacología , Oxazoles/farmacología , Prosencéfalo/metabolismo , Proteínas tau/metabolismo , Animales , Técnicas In Vitro , Masculino , Toxinas Marinas , Fosforilación/efectos de los fármacos , Prosencéfalo/efectos de los fármacos , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteína Fosfatasa 2/metabolismo , Ratas , Ratas Sprague-Dawley , Tirosina/metabolismo
16.
Neuropharmacology ; 52(7): 1521-7, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17434540

RESUMEN

Tau hyperphosphorylation and memory deficit are characteristic alterations of Alzheimer's disease (AD), and glycogen synthase kinase-3 (GSK-3) plays a crucial role in these AD-like changes. We have reported that activation of GSK-3 through ventricular injection of wortmannin and GF-109203X (WT/GFX, 100 microM each) induces tau hyperphosphorylation and memory impairment of rats [Liu, S.J. et al., 2003. Overactivation of glycogen synthase kinase-3 by inhibition of phosphoinositol-3 kinase and protein kinase C leads to hyperphosphorylation of tau and impairment of spatial memory. J. Neurochem. 87, 1333-1344]. By using this model, we explored in the present study the effects of dehydroevodiamine (DHED), a quinazoline alkaloid isolated from Evodia rutaecarpa Bentham, on the memory retention, tau phosphorylation and the activity of GSK-3. We found that pre-administration of DHED through vena caudalis for 1 week efficiently improved the WT/GFX-induced spatial memory retention impairment of the rats; it also antagonized tau hyperphosphorylation at multiple AD sites and arrested the overactivation of GSK-3 induced by WT/GFX. Our study gave the first in vivo evidence that DHED could suppress the overactivation of GSK-3 and improve tau hyperphosphorylation and spatial memory deficit of the rats, suggesting that this chemical may be served as a candidate for arresting AD-like pathological and behavioral alterations.


Asunto(s)
Alcaloides/uso terapéutico , Glucógeno Sintasa Quinasa 3/metabolismo , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/prevención & control , Proteínas tau/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Indoles , Masculino , Maleimidas , Trastornos de la Memoria/inducido químicamente , Fosforilación/efectos de los fármacos , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA