Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Am J Chin Med ; 51(6): 1361-1384, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37489113

RESUMEN

Few studies have reported the therapeutic effects of Korean red ginseng (KRG) against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, the positive effects of KRG on other viruses have been reported and the effects of KRG on pulmonary inflammatory diseases have also been studied. Therefore, this study investigated the therapeutic effects of KRG-water extract (KRG-WE) in a pseudo-type SARS-CoV-2 (PSV)-induced lung injury model. Constructing the pseudovirus, human angiotensin-converting enzyme 2 (hACE2) transgenic mice were infected via intranasal injection that had been orally administered with KRG-WE for six weeks. After 7-days post infection (dpi), the antiviral effects of KRG-WE were confirmed, followed by real-time polymerase chain reaction (PCR), western blot analysis, flow cytometric analysis, and an enzyme-linked immunoassay (ELISA). KRG-WE significantly inhibited an increase in immunoglobulin caused by PSV. Furthermore, KRG-WE effectively suppressed alveolar macrophages (AMs) inside the lungs and helped normalize the population of other immune cells. In addition, virus-induced gene expression and inflammatory signals such as nuclear factor-kappa B and other upstream molecules were downregulated. Moreover, KRG-WE also normalized gene expression and protein activity in the spleen. In conclusion, KRG-WE reduced AMs, normalized the immune response, and decreased the expression of inflammatory genes and activation of signaling pathway phosphorylation, thereby exhibiting anti-inflammatory effects and attenuating lung damage.


Asunto(s)
COVID-19 , Panax , Humanos , Ratones , Animales , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , SARS-CoV-2 , Inflamación/tratamiento farmacológico , Inmunidad
2.
Plants (Basel) ; 12(5)2023 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-36904025

RESUMEN

Ardisia silvestris is a traditional medicinal herb used in Vietnam and several other countries. However, the skin-protective properties of A. silvestris ethanol extract (As-EE) have not been evaluated. Human keratinocytes form the outermost barrier of the skin and are the main target of ultraviolet (UV) radiation. UV exposure causes skin photoaging via the production of reactive oxygen species. Protection from photoaging is thus a key component of dermatological and cosmetic products. In this research, we found that As-EE can prevent UV-induced skin aging and cell death as well as enhance the barrier effect of the skin. First, the radical-scavenging ability of As-EE was checked using DPPH, ABTS, TPC, CUPRAC, and FRAP assays, and a 3-(4-5-dimethylthiazol-2-yl)-2-5-diphenyltetrazolium bromide assay was used to examine cytotoxicity. Reporter gene assays were used to determine the doses that affect skin-barrier-related genes. A luciferase assay was used to identify possible transcription factors. The anti-photoaging mechanism of As-EE was investigated by determining correlated signaling pathways using immunoblotting analyses. As-EE had no harmful effects on HaCaT cells, according to our findings, and As-EE revealed moderate radical-scavenging ability. With high-performance liquid chromatography (HPLC) analysis, rutin was found to be one of the major components. In addition, As-EE enhanced the expression levels of hyaluronic acid synthase-1 and occludin in HaCaT cells. Moreover, As-EE dose-dependently up-regulated the production of occludin and transglutaminase-1 after suppression caused by UVB blocking the activator protein-1 signaling pathway, in particular, the extracellular response kinase and c-Jun N-terminal kinase. Our findings suggest that As-EE may have anti-photoaging effects by regulating mitogen-activated protein kinase, which is good news for the cosmetics and dermatology sectors.

3.
Phytomedicine ; 109: 154573, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36610128

RESUMEN

BACKGROUND: Beauvericin (BEA) is a depsipeptide with antimicrobial, anti-inflammatory, and anti-cancer activities isolated from Beauveria bassiana. However, little is understood on its anti-cancer activities and mechanism. PURPOSE: Aim of this study was to explore the anti-cancer activity of BEA and its underlying molecular mechanism to provide a theoretical basis for its role as a candidate natural drug in cancer diseases. STUDY DESIGN: Various cancer cells such as C6 glioma, U251, MDA-MB-231, HeLa, HCT-15, LoVo cells, and HEK293T cells were used to the anti-cancer activity of BEA. METHODS: To evaluate the anti-cancer activity of BEA, cell viability test (MTT assay), morphological change check, confocal microscopy, actin polymerization assay, flow cytometry, and Western blotting analysis. To check the target enzyme of BEA, overexpression and site-directed mutagenesis was employed. RESULTS: BEA inhibited the viability of cancer cells including C6, MDA-MB-231, HeLa, HCT-15, LoVo, and U251 cells. Treatment of BEA in C6 glioma cells induced cell membrane blebbing and apoptosis. Caspase-3 and -9 were dose-dependently activated by BEA, and the mRNA expression of Bcl-2 was inhibited by BEA. According to confocal microscopy, actin polymerization and actin-actin interaction were interrupted by BEA in C6 cells. BEA regulated the apoptosis of C6 cells depending on the protein phosphorylation of Src and Signal transducer and activator of transcription (STAT3). Moreover, c-terminal amino acids in Src directly interacted with BEA in C6 cells, and the binding of Src and BEA suppressed the kinase activity of Src. CONCLUSIONS: These results suggest that BEA may be a critical candidate or substitute drug for cancer treatment via suppression of the Src/STAT3 pathway.


Asunto(s)
Actinas , Antineoplásicos , Depsipéptidos , Neoplasias , Humanos , Actinas/metabolismo , Apoptosis , Línea Celular Tumoral , Depsipéptidos/farmacología , Células HEK293 , Fosforilación , Polimerizacion , Factor de Transcripción STAT3/metabolismo , Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico
4.
Am J Chin Med ; 51(1): 205-221, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36408728

RESUMEN

20(S)-protopanaxadiol (PPD), a metabolite of Panax ginseng, has multiple pharmacological properties. However, the effects of PPD against human gastric cancer have not been elucidated. Our purpose in this study was to investigate if PPD has anticancer effects against human gastric cancer in vitro. Cell viability, migration, clone formation, and invasion were assessed to explore the effects of PPD on cancer cells. PI and annexin V staining as well as immunoblotting were employed to determine if PPD-induced apoptosis and autophagy of MKN1 and MKN45 cells. The target of PPD was identified using immunoblotting, overexpression analysis, and flow cytometric analysis. PPD exhibited significantly suppressed cell viability, migration, colony formation, and invasion. Phosphorylation of Src and its down-stream effectors were inhibited by PPD. PPD-enhanced apoptosis and autophagy in a dose- and time-dependent manner by inhibiting Src. Collectively, our results demonstrate that PPD induces apoptosis and autophagy in gastric cancer cells in vitro by inhibiting Src.


Asunto(s)
Ginsenósidos , Panax , Sapogeninas , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/tratamiento farmacológico , Apoptosis , Sapogeninas/farmacología , Autofagia , Ginsenósidos/farmacología , Línea Celular Tumoral
5.
Plants (Basel) ; 11(24)2022 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-36559672

RESUMEN

Many species in the genus Guettarda are known to exert anti-inflammatory effects and are used as traditional medicinal plants to treat various inflammatory symptoms. However, no studies on the inflammatory activities of Guettarda crispiflora Vahl have been reported. The aim of the study was to investigate in vitro and in vivo the anti-inflammatory effects of a methanol extract of Guettarda crispiflora Vahl (Gc-ME). To determine the anti-inflammatory activity of Gc-ME, lipopolysaccharide (LPS)-, poly(I:C)-, or Pam3CSK4-treated RAW264.7 cells, HCl/EtOH- and LPS-treated mice were employed for in vitro and in vivo tests. LPS-induced nitric oxide production in RAW264.7 cells was determined by Griess assays and cytokine gene expression in LPS-activated RAW264.7 cells, confirmed by RT- and real-time PCR. Transcriptional activation was evaluated by luciferase reporter gene assay. Target protein validation was assessed by Western blot analysis and cellular thermal shift assays (CETSA) with LPS-treated RAW264.7 and gene-transfected HEK293 cells. Using both a HCl/EtOH-induced gastritis model and an LPS-induced lung injury model, inflammatory states were checked by scoring or evaluating gastric lesions, lung edema, and lung histology. Phytochemical fingerprinting of Gc-ME was observed by using liquid chromatography-mass spectrometry. Nitric oxide production induced by LPS and Pam3CSK4 in RAW264.7 cells was revealed to be reduced by Gc-ME. The LPS-induced upregulation of iNOS, COX-2, IL-6, and IL-1ß was also suppressed by Gc-ME treatment. Gc-ME downregulated the promotor activities of AP-1 and NF-κB triggered by MyD88- and TRIF induction. Upstream signaling proteins for NF-κB activation, namely, p-p50, p-p65, p-IκBα, and p-Src were all downregulated by Ch-EE. Moreover, Src was revealed to be directly targeted by Gc-ME. This extract, orally treated strongly, attenuated the inflammatory symptoms in HCl/EtOH-treated stomachs and LPS-treated lungs. Therefore, these results strongly imply that Guettarda crispiflora can be developed as a promising anti-inflammatory remedy with Src-suppressive properties.

6.
Phytomedicine ; 105: 154338, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35921773

RESUMEN

BACKGROUND: Callerya atropurpurea is a traditional plant in a tropical zone discovered to have anti-inflammatory functions. PURPOSE: we want to investigate the mechanism related to anti-inflammation of C. atropurpurea ethanol extract (Ca-EE) both in vitro and in vivo. STUDY DESIGN: Murine macrophage cells and mouse models for gastritis and septic shock were conducted to evaluate the abilities of Ca-EE in anti-inflammation. METHODS: Ca-EE was tested by HPLC and LC-MS/MS. NO outcome was checked by Griess reagent test. Cell viabilities were evaluated using MTT assay. Inflammatory cytokines were determined via RT-PCR and ELISA. The mechanism of Ca-EE in anti-inflammation was investigated by luciferase reporter gene assay and immunoblot in transcription level and protein level respectively. Gastric injury and septic shock administrated with Ca-EE were studied by H&E, PCR, and immunoblot. RESULTS: Ca-EE significantly decreased LPS-induced NO production, but hardly stimulated the expression of NO itself. It not only showed no cytotoxicity, but also protected cells from LPS damage. Moreover, Ca-EE decreased TLR4 expression, altered MyD88 recruitment and TRAF6, and suppressed the phospho-Src/PI3K/AKT. Ca-EE inhibited downstream signaling P38, JNK and NF-κB. Finally, Ca-EE alleviated HCl/EtOH-induced gastritis and LPS/poly (I:C)-induced septic shock through the previously mentioned signaling cascades. CONCLUSION: Ca-EE exhibited an integrated and promising mechanism against TLR4-related inflammation, which shows potential for treating gastritis, septic shock, and other inflammatory diseases.


Asunto(s)
Fabaceae , Gastritis , Choque Séptico , Animales , Antiinflamatorios , Cromatografía Liquida , Etanol , Inflamación , Lipopolisacáridos , Ratones , Factor 88 de Diferenciación Mieloide , FN-kappa B , Fosfatidilinositol 3-Quinasas , Extractos Vegetales , Espectrometría de Masas en Tándem , Receptor Toll-Like 4
7.
Plants (Basel) ; 11(10)2022 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-35631731

RESUMEN

Growing demand for treatment options against acute lung injury (ALI) emphasizes studies on plant extracts harboring anti-inflammatory effects. According to GC-MS analysis, Angiopteris cochinchinensis de Vriese consists of various flavonoids with anti-inflammatory activities. Thus, in this study, the anti-inflammatory effects of an extract of Angiopteris cochinchinensis de Vriese (Ac-EE) were assessed using RAW264.6 murine macrophages and a lipopolysaccharide (LPS)-induced ALI model. Ac-EE reduced the nitric oxide production in murine macrophages increased by LPS induction. Moreover, protective effects of Ac-EE on lung tissue were demonstrated by shrinkage of edema and lung injury. Reduced neutrophil infiltration and formation of hyaline membranes were also detected in lung tissues after H&E staining. Semiquantitative RT-PCR, quantitative real-time PCR, and ELISA showed that Ac-EE inhibits the production of proinflammatory mediators, including iNOS and COX-2, and cytokines, such as TNF-α, IL-1ß, and IL-6. An Ac-EE-mediated anti-inflammatory response was derived from inhibiting the NF-κB signaling pathway, which was evaluated by luciferase reporter assay and Western blotting analysis. A cellular thermal shift assay revealed that the prime target of Ac-EE in alleviating inflammation was Src. With its direct binding with Src, Angiopteris cochinchinensis de Vriese significantly mitigates lung injury, showing possibilities of its potential as an effective botanical drug.

8.
Molecules ; 26(21)2021 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-34771068

RESUMEN

Caragana rosea Turcz, which belongs to the Leguminosae family, is a small shrub found in Northern and Eastern China that is known to possess anti-inflammatory properties and is used to treat fever, asthma, and cough. However, the underlying molecular mechanisms of its anti-inflammatory effects are unknown. Therefore, we used lipopolysaccharide (LPS) in RAW264.7 macrophages to investigate the molecular mechanisms that underlie the anti-inflammatory activities of a methanol extract of Caragana rosea (Cr-ME). We showed that Cr-ME reduced the production of nitric oxide (NO) and mRNA levels of iNOS, TNF-α, and IL-6 in a concentration-dependent manner. We also found that Cr-ME blocked MyD88- and TBK1-induced NF-κB and IRF3 promoter activity, suggesting that it affects multiple targets. Moreover, Cr-ME reduced the phosphorylation levels of IκBα, IKKα/ß and IRF3 in a time-dependent manner and regulated the upstream NF-κB proteins Syk and Src, and the IRF3 protein TBK1. Upon overexpression of Src and TBK1, Cr-ME stimulation attenuated the phosphorylation of the NF-κB subunits p50 and p65 and IRF3 signaling. Together, our results suggest that the anti-inflammatory activity of Cr-ME occurs by inhibiting the NF-κB and IRF3 signaling pathways.


Asunto(s)
Antiinflamatorios/farmacología , Caragana/química , Inflamación/tratamiento farmacológico , Metanol/química , Extractos Vegetales/farmacología , Animales , Antiinflamatorios/química , Antiinflamatorios/aislamiento & purificación , Células Cultivadas , Células HEK293 , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Factor 3 Regulador del Interferón/antagonistas & inhibidores , Factor 3 Regulador del Interferón/metabolismo , Lipopolisacáridos/antagonistas & inhibidores , Ratones , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/metabolismo
9.
Pharm Biol ; 59(1): 799-810, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34190667

RESUMEN

CONTEXT: Among the plants in the genus Barringtonia (Lecythidaceae) used as traditional medicines to treat arthralgia, chest pain, and haemorrhoids in Indonesia, Barringtonia racemosa L. and Barringtonia acutangula (L.) Gaertn. have demonstrated anti-inflammatory activity in systemic inflammatory models. OBJECTIVE: The anti-inflammatory activity of Barringtonia angusta Kurz has not been investigated. We prepared a methanol extract of the leaves and stems of B. angusta (Ba-ME) and systemically evaluated its anti-inflammatory effects in vitro and in vivo. MATERIALS AND METHODS: RAW264.7 cells stimulated with LPS or Pam3CSK4 for 24 h were treated with Ba-ME (12.5, 25, 50, 100, and 150 µg/mL), and NO production and mRNA levels of inflammatory genes were evaluated. Luciferase reporter gene assay, western blot analysis, overexpression experiments, and cellular thermal shift assay were conducted to explore the mechanism of Ba-ME. In addition, the anti-gastritis activity of Ba-ME (50 and 100 mg/kg, administered twice per day for two days) was evaluated using an HCl/EtOH-induced gastritis mouse model. RESULTS: Ba-ME dose-dependently suppressed NO production [IC50 = 123.33 µg/mL (LPS) and 46.89 µg/mL (Pam3CSK4)] without affecting cell viability. Transcriptional expression of iNOS, IL-1ß, COX-2, IL-6, and TNF-α and phosphorylation of Src, IκBα, p50/105, and p65 were inhibited by Ba-ME. The extract specifically targeted the Src protein by binding to its SH2 domain. Moreover, Ba-ME significantly ameliorated inflammatory lesions in the HCl/EtOH-induced gastritis model. DISCUSSION AND CONCLUSIONS: The anti-inflammatory activity of Ba-ME is mediated by targeting of the Src/NF-κB signalling pathway, and B. angusta has potential as an anti-inflammatory drug.


Asunto(s)
Antiinflamatorios/administración & dosificación , Barringtonia , Sistemas de Liberación de Medicamentos/métodos , Gastritis/tratamiento farmacológico , Extractos Vegetales/administración & dosificación , Familia-src Quinasas/antagonistas & inhibidores , Animales , Antiinflamatorios/aislamiento & purificación , Antiinflamatorios/metabolismo , Relación Dosis-Respuesta a Droga , Gastritis/inducido químicamente , Gastritis/metabolismo , Células HEK293 , Humanos , Masculino , Metanol/administración & dosificación , Metanol/metabolismo , Ratones , Ratones Endogámicos ICR , FN-kappa B , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/metabolismo , Hojas de la Planta , Tallos de la Planta , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Familia-src Quinasas/metabolismo
10.
Molecules ; 25(20)2020 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-33050143

RESUMEN

Muscle atrophy is an abnormal condition characterized by loss of skeletal muscle mass and function and is primarily caused by injury, malnutrition, various diseases, and aging. Leaf of lotus (Nelumbo nucifera Gaertn), which has been used for medicinal purposes, contains various active ingredients, including polyphenols, and is reported to exert an antioxidant effect. In this study, we investigated the effect of water extract of lotus leaf (LL) on muscle atrophy and the underlying molecular mechanisms of action. Amounts of 100, 200, or 300 mg/kg/day LL were administered to dexamethasone (DEX)-induced muscle atrophy mice for 4 weeks. Micro-computed tomography (CT) analysis revealed that the intake of LL significantly increased calf muscle volume, surface area, and density in DEX-induced muscle atrophy mice. Administration of LL recovered moving distance, grip strength, ATP production, and body weight, which were decreased by DEX. In addition, muscle damage caused by DEX was also improved by LL. LL reduced the protein catabolic pathway by suppressing gene expression of muscle atrophy F-Box (MAFbx; atrogin-1), muscle RING finger 1 (MuRF1), and forkhead box O (FoxO)3a, as well as phosphorylation of AMP-activated kinase (AMPK). The AKT-mammalian target of the rapamycin (mTOR) signal pathway, which is important for muscle protein synthesis, was increased in LL-administered groups. The HPLC analysis and pharmacological test revealed that quercetin 3-O-beta-glucuronide (Q3G) is a major active component in LL. Thus, Q3G decreased the gene expression of atrogin-1 and MuRF1 and phosphorylation of AMPK. This compound also increased phosphorylation levels of mTOR and its upstream enzyme AKT in DEX-treated C2C12 cells. We identified that LL improves muscle wasting through regulation of muscle protein metabolism in DEX-induced muscle atrophy mice. Q3G is predicted to be one of the major active phenolic components in LL. Therefore, we propose LL as a supplement or therapeutic agent to prevent or treat muscle wasting, such as sarcopenia.


Asunto(s)
Dexametasona/toxicidad , Lotus/química , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Atrofia Muscular/tratamiento farmacológico , Atrofia Muscular/metabolismo , Extractos Vegetales/uso terapéutico , Hojas de la Planta/química , Agua/química , Animales , Western Blotting , Línea Celular , Cromatografía Líquida de Alta Presión , Masculino , Ratones , Extractos Vegetales/química , Reacción en Cadena en Tiempo Real de la Polimerasa , Microtomografía por Rayos X
11.
Biomolecules ; 10(4)2020 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-32331432

RESUMEN

Inflammation is a complex protective response of body tissues to harmful stimuli. Acute inflammation can progress to chronic inflammation, which can lead to severe disease. Therefore, this research focuses on the development of anti-inflammatory drugs, and natural extracts have been explored as potential agents. No study has yet examined the inflammation-associated pharmacological activity of Potentilla glabra Var. mandshurica (Maxim.) Hand.-Mazz ethanol extract (Pg-EE). To examine the mechanisms by which Pg-EE exerts anti-inflammatory effects, we studied its activities in lipopolysaccharide (LPS)-treated murine macrophage RAW264.7 cells and an HCl/EtOH-induced gastritis model. LPS-triggered nitric oxide (NO) release and mRNA levels of inducible nitric oxide synthase (iNOS), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1 beta (IL-1ß) in RAW264.7 cells were suppressed by Pg-EE in a dose-dependent manner. Using a luciferase assay and western blot assay, we found that the NF-κB pathway was inhibited by Pg-EE, particularly by the decreased level of phosphorylated proteins of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) subunits (p65 and p50), inhibitor of kappa B alpha (IκBα), p85, and Src. Using an overexpression strategy, cellular thermal shift assay, and immunoprecipitation analysis, we determined that the anti-inflammatory effect of Pg-EE was mediated by the inhibition of Src. Pg-EE further showed anti-inflammatory effects in vivo in the HCl/EtOH-induced gastritis mouse model. In conclusion, Pg-EE exerts anti-inflammatory activities by targeting Src in the NF-κB pathway, and these results suggest that Pg-EE could be used as an anti-inflammatory herbal medicine.


Asunto(s)
Antiinflamatorios/farmacología , Etanol/química , FN-kappa B/antagonistas & inhibidores , Extractos Vegetales/farmacología , Potentilla/química , Familia-src Quinasas/antagonistas & inhibidores , Enfermedad Aguda , Animales , Gastritis/patología , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Modelos Biológicos , FN-kappa B/metabolismo , Óxido Nítrico/biosíntesis , Células RAW 264.7 , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcripción Genética/efectos de los fármacos , Familia-src Quinasas/metabolismo
12.
J Ethnopharmacol ; 235: 38-46, 2019 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-30710734

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Olea europaea L., (Oleaceae) has been used widely in folk medicine in the European Mediterranean islands, India, Asia, and other parts of the world. Although this plant has high ethnopharmacological value for treating inflammatory diseases, the molecular mechanisms of how it inhibits the inflammatory response are not fully understood. In this study, we sought to identify the anti-inflammatory mechanisms of this plant. MATERIALS AND METHODS: Using macrophages, we investigated the effects of O. europaea L. methanol extract (Oe-ME) and ethanol extract (Oe-EE) on the production of inflammatory mediator nitric oxide (NO) and prostaglandin E2 (PGE2), the expression levels of pro-inflammatory genes and intracellular inflammatory signaling activities. RESULTS: Oe-ME and Oe-EE suppressed the production of NO in lipopolysaccharide-(LPS-), Pam3CSK4-, and poly (I:C)-stimulated RAW264.7 cells; importantly, no cytotoxicity was observed. Oe-ME and Oe-EE reduced production of PGE2 without exhibiting cytotoxicity. The mRNA expression levels of cyclooxygenase-2 (COX-2), inducible NO synthase (iNOS), IL-6, IL-1ß, and tumor necrosis factor (TNF)-α were down-regulated by Oe-ME and Oe-EE. Nuclear fraction and whole lysate immunoblotting analyses and overexpression experiments strongly suggested that Oe-ME decreased the translocation of p65 and p50 (nuclear factors of the NF-κB subunit) as well as Src and Syk. CONCLUSION: These results suggest that Oe-ME exerts its anti-inflammatory effects by targeting Src and Syk in the NF-κB signaling pathway.


Asunto(s)
Antiinflamatorios/farmacología , Inflamación/tratamiento farmacológico , Olea/química , Extractos Vegetales/farmacología , Animales , Antiinflamatorios/aislamiento & purificación , Dinoprostona/metabolismo , Etanol/química , Células HEK293 , Humanos , Inflamación/patología , Mediadores de Inflamación/metabolismo , Macrófagos/efectos de los fármacos , Metanol/química , Ratones , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Quinasa Syk/metabolismo , Familia-src Quinasas/metabolismo
13.
Phytother Res ; 33(3): 676-689, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30632216

RESUMEN

Although Morinda citrifolia (noni) has long been used in traditional medicines for human diseases, its molecular and cellular mechanism of immunostimulatory ability to improve human health under normal healthy conditions is not fully elucidated. This study aimed to investigate the in vitro and in vivo immunostimulatory activity of M. citrifolia fruit water extract treated with enzymes (Mc-eWE). In vitro studies revealed that Mc-eWE stimulated the cells by inducing nitric oxide (NO) production and the expression of inflammatory cytokines, such as interleukin (IL)-1ß, IL-6, IL-12, tumor necrosis factor-alpha (TNF-α), and interferon-gamma (IFN-γ). The immunostimulatory activity was mediated by activation of NF-κB and AP-1. Ex vivo studies showed that Mc-eWE stimulated splenocytes isolated from mice by inducing NO production and expression of immunostimulatory cytokines and by downregulating the expression of the immunosuppressive cytokine IL-10 without cytotoxicity. In vivo demonstrated that Mc-eWE induced immunostimulation by modulating populations of splenic immune cells, especially by increasing the population of IFN-γ+ NK cells. Mc-eWE enhanced the expression of inflammatory genes and immunostimulatory cytokines and inhibited the expression of IL-10 in the mouse splenocytes and sera. Taken together, these results suggest that Mc-eWE plays an immunostimulatory role by activating innate and adaptive immune responses.


Asunto(s)
Morinda , Extractos Vegetales/farmacología , Inmunidad Adaptativa/efectos de los fármacos , Adyuvantes Inmunológicos/farmacología , Animales , Citocinas/análisis , Inmunidad Innata/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/biosíntesis , Células RAW 264.7
14.
J Ethnopharmacol ; 231: 1-9, 2019 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-30415059

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Mycetia cauliflora Reinw. (Rubiaceae) has been used as a traditional remedy to ameliorate clinical signs of inflammatory diseases, including pain, inflammation, ulcers, and wounds. Among the Mycetia subfamilies, the molecular and cellular mechanisms of Mycetia longifolia (Rubiaceae) have been studied. However, those of Mycetia cauliflora are not clearly understood. Comprehensive investigation of this plant is necessary to evaluate its potential for ethnopharmacological use. MATERIALS: and methods: The activities of Mycetia cauliflora methanol extract (Mc-ME) on the secretion of inflammatory mediators, the mRNA expression of proinflammatory cytokines, and identification of its molecular targets were elucidated using lipopolysaccharide (LPS)-induced macrophage-like cells. Moreover, the suppressive actions of Mc-ME were examined in an LPS-induced peritonitis mouse model. RESULTS: At nontoxic concentrations, Mc-ME downregulated the release of nitric oxide (NO), the mRNA expression of inducible nitric oxide synthase (iNOS), and the mRNA expression of interleukin (IL)-1ß from LPS-activated RAW264.7 cells. This extract also inhibited the nuclear translocation of p65 and p50 and the phosphorylation of IκBα, IKK, and AKT. Western blot analysis and in vitro kinase assays confirmed that phosphoinositide-dependent kinase-1 (PDK1) is the direct immunopharmacological target of Mc-ME effect. In addition, Mc-ME significantly reduced inflammatory signs in an animal model of acute peritonitis. These effects were associated with decreased NO production and decreased AKT phosphorylation. CONCLUSION: Our results suggest that Mc-ME displays anti-inflammatory actions in LPS-treated macrophage-like cells and in an animal model of acute inflammatory disease. These actions are preferentially managed by targeting PDK1 in the nuclear factor (NF)-κB signaling pathway.


Asunto(s)
Antiinflamatorios/farmacología , Extractos Vegetales/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Rubiaceae , Animales , Antiinflamatorios/uso terapéutico , Células HEK293 , Humanos , Interleucina-1beta/genética , Lipopolisacáridos , Macrófagos Peritoneales/efectos de los fármacos , Masculino , Metanol/química , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Peritonitis/inducido químicamente , Peritonitis/tratamiento farmacológico , Peritonitis/metabolismo , Extractos Vegetales/uso terapéutico , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Solventes/química
15.
Artículo en Inglés | MEDLINE | ID: mdl-29725354

RESUMEN

Celtis choseniana is the traditional plant used at Korea as a herbal medicine to ameliorate inflammatory responses. Although Celtis choseniana has been traditionally used as a herbal medicine at Korea, no systemic research has been conducted on its anti-inflammatory activity. Therefore, the present study explored an anti-inflammatory effect and its underlying molecular mechanism using Celtis choseniana methanol extract (Cc-ME) in macrophage-mediated inflammatory responses. In vitro anti-inflammatory activity of Cc-ME was evaluated using RAW264.7 cells and peritoneal macrophages stimulated by lipopolysaccharide (LPS), pam3CSK4 (Pam3), or poly(I:C). In vivo anti-inflammatory activity of Cc-ME was investigated using acute inflammatory disease mouse models, such as LPS-induced peritonitis and HCl/EtOH-induced gastritis. The molecular mechanism of Cc-ME-mediated anti-inflammatory activity was examined by Western blot analysis and immunoprecipitation using whole cell and nuclear fraction prepared from the LPS-stimulated RAW264.7 cells and HEK293 cells. Cc-ME inhibited NO production and mRNA expression of inducible nitric oxide synthase (iNOS), cyclooxygenase (COX-2), and tumor necrosis factor-alpha (TNF-α) in the RAW264.7 cells and peritoneal macrophages induced by LPS, pam3, or poly(I:C) without cytotoxicity. High-performance liquid chromatography (HPLC) analysis showed that Cc-ME contained anti-inflammatory flavonoids quercetin, luteolin, and kaempferol. Among those, the content of luteolin, which showed an inhibitory effect on NO production, was highest. Cc-ME suppressed the NF-κB signaling pathway by targeting Src and interrupting molecular interactions between Src and p85, its downstream kinase. Moreover, Cc-ME ameliorated the morphological finding of peritonitis and gastritis in the mouse disease models. Therefore, these results suggest that Cc-ME exerted in vitro and in vivo anti-inflammatory activity in LPS-stimulated macrophages and mouse models of acute inflammatory diseases. This anti-inflammatory activity of Cc-ME was dominantly mediated by targeting Src in NF-κB signaling pathway during macrophage-mediated inflammatory responses.

16.
J Ethnopharmacol ; 219: 202-212, 2018 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-29574093

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Alisma canaliculatum A.Braun & C.D.Bouché, distributed in Korea, Japan, China, and Taiwan, is a traditional medicine. In particular, the stem and root of Alisma canaliculatum A.Braun & C.D.Bouché are prescribed to relieve various inflammatory symptoms resulting from nephritis, cystitis, urethritis, and dropsy. AIM OF STUDY: However, the curative mechanism of Alisma canaliculatum A.Braun & C.D.Bouché with respect to inflammatory symptoms is poorly understood. In this study, the curative roles of this plant in various inflammatory conditions as well as its inhibitory mechanism were aimed to examine using an ethanol extract (Ac-EE). MATERIALS AND METHODS: Anti-inflammatory effects of Ac-EE were evaluated in lipopolysaccharide (LPS)-induced macrophages in vitro and HCl/EtOH-stimulated mouse model of gastritis and DSS-treated mouse model of colitis. To determine the potentially active anti-inflammatory components in this extracts, we employed HPLC. We also used kinase assays, reporter gene assay, immunoprecipitation analysis and target enzyme overexpressing cell analysis to analyze the molecular mechanisms and the target molecules. RESULTS: This extract dose-dependently inhibited the production of nitric oxide (NO) and prostaglandin E2 (PGE2) from RAW264.7 cells and peritoneal macrophages activated by lipopolysaccharide (LPS). Additionally, Ac-EE ameliorated inflammatory symptoms resulting from gastritis and colitis. Ac-EE down-regulated the mRNA levels of inducible NO synthase (iNOS), tumor necrosis factor (TNF)-α, and cyclooxygenase-2 (COX-2). Ac-EE also blocked the nuclear translocation of nuclear factor (NF)-κB and activator protein (AP)- 1 in LPS-stimulated RAW264.7 cells. By analyzing the target signaling molecules activating these transcription factors, we found that Src and Syk, as well as molecular association between TAK1 and mitogen-activated protein kinase kinase 4/7 (MKK4/7), were targeted by Ac-EE. CONCLUSIONS: Our data suggest that the Ac-EE NF-κB/AP-1-targeted anti-inflammatory potential is mediated by suppression of Src and Syk as well as the complex formation between TAK1 and its substrate proteins MKK4/7.


Asunto(s)
Alisma , Antiinflamatorios/uso terapéutico , Gastritis/tratamiento farmacológico , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Extractos Vegetales/uso terapéutico , Quinasa Syk/antagonistas & inhibidores , Familia-src Quinasas/antagonistas & inhibidores , Animales , Antiinflamatorios/aislamiento & purificación , Antiinflamatorios/farmacología , Sulfato de Dextran/toxicidad , Etanol/toxicidad , Gastritis/inducido químicamente , Gastritis/metabolismo , Células HEK293 , Humanos , Ácido Clorhídrico/toxicidad , Lipopolisacáridos/toxicidad , Quinasas Quinasa Quinasa PAM/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/farmacología , Células RAW 264.7 , Quinasa Syk/metabolismo , Familia-src Quinasas/metabolismo
17.
Artículo en Inglés | MEDLINE | ID: mdl-30598682

RESUMEN

Abutilon crispum L. Medik, better known as bladdermallow, is used as a traditional remedy in India, for its anti-inflammatory effect due to its high content of flavonoids. However, research about its anti-inflammatory effect at the molecular level has not been performed. In this study, we aimed to investigate the mechanism of Abutilon crispum methanol extract (Ac-ME) in inhibiting the inflammatory response by conducting several experiments including cellular and molecular assays. Ac-ME inhibited the production of nitric oxide (NO) in RAW264.7 cells during treatment of LPS and Pam3CSK4 without exhibiting cytotoxicity. Ac-ME also suppressed the mRNA expression of inducible nitric oxide (iNOS) and proinflammatory cytokines such as interleukin (IL)-1ß and IL-6. Moreover, Ac-ME was shown to inhibit the NF-κB pathway, according to the luciferase reporter gene assay performed with a NF-κB-Luc construct containing NF-κB-binding promoter regions under MyD88 and TRIF overexpression conditions, and immunoblotting analysis by determining the phospho-form levels of IκBα, IKKα/ß, and p85, a regulatory domain of phosphatidylinositide 3-kinase (PI3K). Finally, we observed that the level of phospho-p85 induced by the overexpression of spleen tyrosine kinase (Syk) and Src was decreased by Ac-ME at 200 µg/ml. Therefore, these results suggest that Ac-ME has an anti-inflammatory effect by targeting PI3K in the NF-κB signaling pathway.

18.
J Ethnopharmacol ; 206: 1-7, 2017 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-28502904

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Nuclear factor-kappa B (NF-κB) plays pivotal roles in inflammation. Src and Syk are two tyrosine kinases that act upstream of NF-κB signaling. Although Achyranthes aspera L. (A. aspera) has been used as a traditional medicine to treat fevers and inflammatory ailments and heal wounds, the molecular mechanisms of its anti-inflammatory actions are not yet fully understood. MATERIALS AND METHODS: In this study, we evaluated the anti-inflammatory effect of A. aspera ethanol extract (Aa-EE). To determine the mechanism by which Aa-EE dampens the inflammatory response, nitric oxide (NO) production and the mRNA expression levels of tumor necrosis factor (TNF)-α and inducible nitric oxide synthase (iNOS) were examined by Griess assay and RT-PCR. Luciferase assays and immunoblotting were also conducted to examine how Aa-EE regulates the NF-κB pathway. RESULTS: Aa-EE reduced NO production up to 60% without any cytotoxicity. This extract was found to downregulate the mRNA expression levels of inflammatory genes. Aa-EE blocked NF-κB promoter activity induced by both TNF-α and adaptor molecule MyD88 (about 70% and 40%, respectively). Moreover, nuclear translocation of p65 and IκBα phosphorylation were also inhibited. Furthermore, Aa-EE inactivated two upstream signaling molecules, the Src and Syk kinases. In accordance with these data, the kinase activities of Src and Syk were decreased by 50% and 80%, respectively. The anti-inflammatory action of Aa-EE was also confirmed in a gastritis model. CONCLUSION: Our data suggest that Aa-EE targets NF-κB to exert its anti-inflammatory properties by suppressing Src and Syk. Therefore, our study raises the possibility that this extract can be developed as a novel natural anti-inflammatory remedy.


Asunto(s)
Achyranthes/química , Antiinflamatorios/farmacología , Extractos Vegetales/farmacología , Quinasa Syk/metabolismo , Familia-src Quinasas/metabolismo , Animales , Línea Celular , Etanol , Humanos , Masculino , Ratones , Ratones Endogámicos ICR
19.
Am J Chin Med ; 44(3): 595-615, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27109153

RESUMEN

Korean Red Ginseng (KRG) is an herbal medicine prescribed worldwide that is prepared from Panax ginseng C.A. Meyer (Araliaceae). Out of ginseng's various components, ginsenosides are regarded as the major ingredients, exhibiting anticancer and anti-inflammatory activities. Although recent studies have focused on understanding the anti-inflammatory activities of KRG, compounds that are major anti-inflammatory components, precisely how these can suppress various inflammatory processes has not been fully elucidated yet. In this study, we aimed to identify inhibitory saponins, to evaluate the in vivo efficacy of the saponins, and to understand the inhibitory mechanisms. To do this, we employed in vitro lipopolysaccharide-treated macrophages and in vivo inflammatory mouse conditions, such as collagen (type II)-induced arthritis (CIA), EtOH/HCl-induced gastritis, and lipopolysaccharide (LPS)/D-galactosamine (D-GalN)-triggered hepatitis. Molecular mechanisms were also verified by real-time PCR, immunoblotting analysis, and reporter gene assays. Out of all the ginsenosides, ginsenoside Rc (G-Rc) showed the highest inhibitory activity against the expression of tumor necrosis factor (TNF)-[Formula: see text], interleukin (IL)-1[Formula: see text], and interferons (IFNs). Similarly, this compound attenuated inflammatory symptoms in CIA, EtOH/HCl-mediated gastritis, and LPS/D-galactosamine (D-GalN)-triggered hepatitis without altering toxicological parameters, and without inducing gastric irritation. These anti-inflammatory effects were accompanied by the suppression of TNF-[Formula: see text] and IL-6 production and the induction of anti-inflammatory cytokine IL-10 in mice with CIA. G-Rc also attenuated the increased levels of luciferase activity by IRF-3 and AP-1 but not NF-[Formula: see text]B. In support of this phenomenon, G-Rc reduced TBK1, IRF-3, and ATF2 phosphorylation in the joint and liver tissues of mice with hepatitis. Therefore, our results strongly suggest that G-Rc may be a major component of KRG with useful anti-inflammatory properties due to its suppression of IRF-3 and AP-1 pathways.


Asunto(s)
Artritis/tratamiento farmacológico , Gastritis/tratamiento farmacológico , Ginsenósidos/administración & dosificación , Hepatitis/tratamiento farmacológico , Panax/química , Fitoterapia , Factor de Transcripción Activador 2 , Administración Oral , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ginsenósidos/aislamiento & purificación , Ginsenósidos/farmacología , Células HEK293 , Humanos , Mediadores de Inflamación/metabolismo , Factor 3 Regulador del Interferón , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos ICR , Células RAW 264.7
20.
Artículo en Inglés | MEDLINE | ID: mdl-25878717

RESUMEN

In traditional Chinese medicine, Persicaria chinensis L. has been prescribed to cure numerous inflammatory disorders. We previously analyzed the bioactivity of the methanol extract of this plant (Pc-ME) against LPS-induced NO and PGE2 in RAW264.7 macrophages and found that it prevented HCl/EtOH-induced gastric ulcers in mice. The purpose of the current study was to explore the molecular mechanism by which Pc-ME inhibits activator protein- (AP-) 1 activation pathway and mediates its hepatoprotective activity. To investigate the putative therapeutic properties of Pc-ME against AP-1-mediated inflammation and hepatotoxicity, lipopolysaccharide- (LPS-) stimulated RAW264.7 and U937 cells, a monocyte-like human cell line, and an LPS/D-galactosamine- (D-GalN-) induced acute hepatitis mouse model were employed. The expression of LPS-induced proinflammatory cytokines including interleukin- (IL-) 1ß, IL-6, and tumor necrosis factor-α (TNF-α) was significantly diminished by Pc-ME. Moreover, Pc-ME reduced AP-1 activation and mitogen-activated protein kinase (MAPK) phosphorylation in both LPS-stimulated RAW264.7 cells and differentiated U937 cells. Additionally, we highlighted the hepatoprotective and curative effects of Pc-ME pretreated orally in a mouse model of LPS/D-GalN-intoxicated acute liver injury by demonstrating the significant reduction in elevated serum AST and ALT levels and histological damage. Therefore, these results strongly suggest that Pc-ME could function as an antihepatitis remedy suppressing MAPK/AP-1-mediated inflammatory events.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA