Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Ethnopharmacol ; 329: 118140, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38565409

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Qingfu Juanbi Tang (QFJBT), a novel and improved Chinese herbal formulation, has surged in recent years for its potential in the therapy of rheumatoid arthritis (RA). Anti-arthritic effects and underlying molecular mechanisms of QFJBT have increasingly become a focal point in research. AIM OF THE STUDY: This study utilized network pharmacology, molecular docking, and experimental validation to elucidate effective ingredients and anti-arthritic mechanisms of QFJBT. MATERIALS AND METHODS: Targets associated with QFJBT and RA were identified from relevant databases and standardized using the Uniprot for gene nomenclature. A "QFJBT-ingredient-target network" and a "Venn diagram of QFJBT and RA targets" were created from the data. The overlap in the Venn diagram highlighted potential targets of QFJBT in the treatment of RA. These targets were subjected to PPI network, GO, and KEGG pathway analysis. The findings were subsequently confirmed through molecular docking and pharmacological experiments to propose the mechanism of action of QFJBT. RESULTS: The study identified 236 active ingredients in QFJBT, with 120 predicted to be effective against RA. Molecular docking showed high binding affinity of key targets (JUN, PTGS2, and TNF-α) with bioactive compounds (rhein, sinomenine, calycosin, and paeoniflorin) of QFJBT. Pharmacodynamic evaluation demonstrated the effects of QFJBT at the dose of 4.56 g/kg in ameliorating symptoms of AIA rats and in reducing levels of JUN, PTGS2, and TNF-α in synovial tissues. In vitro studies further exhibited that rhein, paeoniflorin, sinomenine, calycosin, and QFJBT-containing serum significantly inhibited abnormal proliferation of RA fibroblast-like synoviocytes. Interestingly, rhein and paeoniflorin specifically decreased p-JUN/JUN expression and TNF-α release, respectively, while sinomenine and calycosin selectively increased PTGS2 expression. Consistently, QFJBT-containing serum demonstrated similar effects as those active ingredients identified in QFJBT did. CONCLUSIONS: QFJBT, QFJBT-containing serum, and its active ingredients (rhein, paeoniflorin, sinomenine, and calycosin) suppress inflammatory responses in RA. Anti-arthritic effects of QFJBT and its active ingredients are likely linked to their modulatory impact on identified hub targets.


Asunto(s)
Antirreumáticos , Artritis Reumatoide , Ciclooxigenasa 2 , Medicamentos Herbarios Chinos , Simulación del Acoplamiento Molecular , Animales , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Antirreumáticos/farmacología , Antirreumáticos/uso terapéutico , Ratas , Masculino , Ciclooxigenasa 2/metabolismo , Farmacología en Red , Ratas Sprague-Dawley , Sinoviocitos/efectos de los fármacos , Sinoviocitos/metabolismo , Morfinanos/farmacología , Morfinanos/uso terapéutico , Morfinanos/química , Artritis Experimental/tratamiento farmacológico , Humanos , Descubrimiento de Drogas/métodos
2.
Pharmacology ; 109(2): 76-85, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38290489

RESUMEN

BACKGROUND: Cancer is a major cause of death worldwide. Although modern medicine has made strides in treatment, a complete cure for cancer remains elusive. SUMMARY: Utilization of medicinal plants in traditional medicine for the treatment of multiple diseases, including cancer, is a well-established practice. Sinomenine is an alkaloid extracted from a medicinal plant and has a diverse range of biological properties, including anti-oxidative, anti-inflammatory, and antibacterial effects. Sinomenine exhibits inhibitory effects on various types of tumor cells, including breast, lung, and liver cancers. The anticancer properties of sinomenine are believed to involve stimulation of apoptosis and autophagy as well as suppression of cell proliferation, invasion, and metastasis. KEY MESSAGE: This review summarizes the current research on sinomenine's potential as an anticancer agent, which may contribute to the discovery of more effective cancer treatments.


Asunto(s)
Antineoplásicos , Morfinanos , Neoplasias , Plantas Medicinales , Antiinflamatorios , Morfinanos/farmacología , Morfinanos/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico
3.
Fitoterapia ; 172: 105713, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37949304

RESUMEN

The chemical structure of sinoacutine is formed by a phenanthrene nucleus and an ethylamine bridge. Because it has a similar parent structure to morphine, it is subdivided into morphinane. At present, all reports have pointed out that the basic skeleton of morphine alkaloids is salutaridine (the isomer of sinoacutine), which is generated by the phenol coupling reaction of (R)-reticuline. This study shows that the biosynthetic precursors of sinoacutine and salutaridine are different. In this paper, the sinoacutine synthetase (SinSyn) gene was cloned from Sinomenium acutum and expressed SinSyn protein. Sinoacutine was produced by SinSyn catalyzed (S)-reticuline, according to the results of enzyme-catalyzed experiments. The optical activity, nuclear magnetic resonance, and mass spectrum of sinoacutine and salutaridine were analyzed. The classification and pharmacological action of isoquinoline alkaloids were discussed. It was suggested that sinoacutine should be separated from morphinane and classified as sinomenine alkaloids.


Asunto(s)
Alcaloides , Morfinanos , Estructura Molecular , Morfinanos/química , Morfinanos/metabolismo , Morfinanos/farmacología , Alcaloides/farmacología , Derivados de la Morfina
4.
Bioorg Med Chem Lett ; 97: 129545, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37939862

RESUMEN

Traditional Chinese medicine Qingfengteng primarily acquired from the dried canes of Sinomenium acutum (Thunb.) Rehd. et Wils. var. cinereum Rehd. et Wils. and S. acutum (Thunb.) Rehd. et Wils. For the therapeutic treatment of rheumatism, acute arthritis, and rheumatoid arthritis based on Qingfengteng, sinomenine hydrochloride was recently made the principal active ingredient in various dosage forms. 8-Bis(benzylthio)octanoic acid (CPI-613) was an orphan medicine that the FDA and EMA approved orphan for the treatment of certain resistant malignancies. Its unique mode of action and minimal toxicity toward normal tissues made for an apt pharmacophore. In order to expand the field of sinomenine anticancer structures, sinomenine/8-Bis(benzylthio)octanoic acid derivatives were designed and synthesized. Among them, target hybrids e4 stood out for having notable cytotoxic effects against cancer cell lines, especially for K562 cells, with IC50 values of 2.45 µM and high safety. In-depth investigations demonstrated that e4 caused apoptosis by stopping the cell cycle at G1 phase, and doing so by altering the morphology of the nucleus and causing membrane potential of the in mitochondria to collapse. These results indicated e4 exerted an antiproliferative effect through apoptosis induction via mitochondrial pathway.


Asunto(s)
Morfinanos , Caprilatos/farmacología , Medicina Tradicional China , Morfinanos/farmacología , Morfinanos/química
5.
Phytomedicine ; 121: 155114, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37816287

RESUMEN

BACKGROUND: Sinomenine (SIN) is the main pharmacologically active component of Sinomenii Caulis and protects against rheumatoid arthritis (RA). In recent years, many studies have been conducted to elucidate the pharmacological mechanisms of SIN in the treatment of RA. However, the molecular mechanism of SIN in RA has not been fully elucidated. PURPOSE: To summarize the pharmacological effects and molecular mechanisms of SIN in RA and clarify the most valuable regulatory mechanisms of SIN to provide clues and a basis for basic research and clinical applications. METHODS: We systematically searched SciFinder, Web of Science, PubMed, China National Knowledge Internet (CNKI), the Wanfang Databases, and the Chinese Scientific Journal Database (VIP). We organized our work based on the PRISMA statement and selected studies for review based on predefined selection criteria. OUTCOME: After screening, we identified 201 relevant studies, including 88 clinical trials and 113 in vivo and in vitro studies on molecular mechanisms. Among these studies, we selected key results for reporting and analysis. CONCLUSIONS: We found that most of the known pharmacological mechanisms of SIN are indirect effects on certain signaling pathways or proteins. SIN was manifested to reduce the release of inflammatory cytokines such as Tumor necrosis factor-α (TNF-α), Interleukin-6 (IL-6), and IL-1ß, thereby reducing the inflammatory response, and apparently blocking the destruction of bone and cartilage. The regulatory effects on inflammation and bone destruction make SIN a promising drug to treat RA. More notably, we believe that the modulation of α7nAChR and the regulation of methylation levels at specific GCG sites in the mPGES-1 promoter by SIN, and its mechanism of directly targeting GBP5, certainly enriches the possibilities and the underlying rationale for SIN in the treatment of inflammatory immune-related diseases.


Asunto(s)
Artritis Reumatoide , Morfinanos , Humanos , Artritis Reumatoide/tratamiento farmacológico , Antiinflamatorios/farmacología , Morfinanos/farmacología , Morfinanos/uso terapéutico , Transducción de Señal
6.
Naunyn Schmiedebergs Arch Pharmacol ; 396(11): 2943-2955, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37133789

RESUMEN

Haematocarpus validus (Miers) Bakh. f. ex Forman, a lesser-known fruit and medicinal plant of high nutraceutical and medicinal value, is used as anti-arthritic, hepatoprotective, and anti-inflammatory agents in ethnomedicine. Metabolome studies in H. validus are a virgin area of research and here we report the spectra of non-volatiles present in the methanolic leaf and fruit extract, using high-resolution liquid chromatography-mass spectrometry. Furthermore, the alkaloid sinomenine was quantified using high-performance thin layer chromatography spectrodensitometric analysis owing to its pharmacological importance as anti-arthritic and anti-inflammatory drug. Electrospray ionization with protonation in positive mode was selected for the analysis and the spectral data was interrogated using MassHunter software. A total of 40 compounds were identified from leaf and fruit samples and the major classes of compounds identified were alkaloids, terpenoids, steroids, tripeptides, vitamins, and related compounds. For separation and quantitation of sinomenine, chloroform:methanol:water (60:30:6.5, v/v) was used as the mobile phase and sinomenine hydrochloride as reference compound. The analysis confirmed the presence of sinomenine in both non-defatted and defatted methanolic leaf extract with quantities 45.73 and 26.02 mg/100 g dry weight, respectively. H. validus is a non-conventional source of sinomenine, the anti-arthritic and anti-inflammatory alkaloid. Sinomenine detected in this study supports the ethnomedicinal uses of H. validus as an anti-arthritic agent. Further study is needed to elucidate the underlying molecular mechanism of its anti-arthritic attributes as well as the corresponding structure-activity relationships.


Asunto(s)
Alcaloides , Antineoplásicos , Morfinanos , Frutas , Morfinanos/farmacología , Antiinflamatorios/uso terapéutico , Metanol , Extractos Vegetales/uso terapéutico , Extractos Vegetales/farmacología
7.
Yakugaku Zasshi ; 143(2): 153-158, 2023.
Artículo en Japonés | MEDLINE | ID: mdl-36724928

RESUMEN

Sinomenine (SIN) is a major component contained in extracts of the Chinese medicinal herb Sinomenium acutum. SIN has various pharmacological properties, including cytoprotection, immunosuppression and anti-inflammation effects. Furthermore, recent studies have reported that SIN has anti-tumor and antidepressant effects, which has created a strong need for SIN kinetic studies. This paper reports a simple and sensitive competitive enzyme-linked immunosorbent assay (ELISA) for the pharmacokinetic evaluation of SIN. Anti-SIN serum was obtained by immunizing mice with an antigen conjugated with bovine serum albumin and carboxylic modified SIN using the N-succinimidyl ester method. Enzyme labeling of SIN with horseradish peroxidase was similarly performed using carboxylic modified SIN. Under optimized conditions, this ELISA shows a linear detection range from 40 to 5000 pg/mL, and a limit of detection of 12.1 pg/mL for 50-µL samples. This assay was specific for SIN and showed very slight cross-reactivity with dextromethorphan (0.45%), dimemorfan (0.22%) and codeine (0.01%), but no cross-reactivity with 2-methoxycyclohex-2-enone (<0.001%). Using this ELISA, SIN levels were easily determined in the blood of mice after oral administration of Kampo medicine, Boiogito. The ELISA may be a valuable tool for studies of the biological and pharmacological properties of SIN.


Asunto(s)
Morfinanos , Ratones , Animales , Cinética , Ensayo de Inmunoadsorción Enzimática/métodos , Morfinanos/farmacología , Antígenos
8.
J Ethnopharmacol ; 303: 116037, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36526094

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: The rhizome of Chinese medical plant QingTeng (scientific name: Sinomenium acutum (Thunb.) Rehd. et Wils.) is widely used by traditional medical doctors for anti-inflammation and immunoregulatory in China and other Asian countries. AIM OF THE STUDY: The purpose of this study was to evaluate the effects and possible mechanisms of sinomenine resistance against DSS-induced inflammation in vitro and in vivo. MATERIALS AND METHODS: The UC model was induced by treating female mice with 3% DSS in vivo and human colonic epithelial cells (Hcoepic) with 0.8 mg/ml DSS in vitro. The mice and Hcoepic were then treated with sinomenine. Inflammatory factors were detected using ELISA and qRT-PCR. Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) p65 and 14-3-3θ were analyzed by bioinformatic analysis and verified by western blotting, immunofluorescent staining or immunohistochemistry. RESULTS: DSS-induced Hcoepic underwent high inflammation and oxidative stress conditions, whereas sinomenine reduced the uncontrolled immune microenvironment by suppressing NF-κB signaling and targeting 14-3-3θ. Knockdown of 14-3-3θ decreased the protective effect of sinomenine against DSS-induced inflammation in vitro. Moreover, sinomenine promoted 14-3-3θ protein expression and inhibited NF-κB p65 signaling in DSS-induced mice. CONCLUSION: These findings suggest that 14-3-3θ plays an important role in sinomenine against DSS treatment, and sinomenine could be considered a potential drug for the treatment of UC.


Asunto(s)
Colitis Ulcerosa , Morfinanos , Animales , Humanos , Ratones , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/tratamiento farmacológico , Sulfato de Dextran , Modelos Animales de Enfermedad , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Ratones Endogámicos C57BL , Morfinanos/farmacología , FN-kappa B/metabolismo , Transducción de Señal
9.
Molecules ; 27(24)2022 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-36557779

RESUMEN

Sinomenine is the main component of the vine Sinomenium acutum. It was first isolated in the early 1920s and has since attracted special interest as a potential anti-rheumatoid arthritis (RA) agent, owing to its successful application in traditional Chinese medicine for the treatment of neuralgia and rheumatoid diseases. In the past few decades, significant advances have broadened our understanding of the molecular mechanisms through which sinomenine treats RA, as well as the structural modifications necessary for improved pharmacological activity. In this review, we summarize up-to-date reports on the pharmacological properties of sinomenine in RA treatment, document their underlying mechanisms, and provide an overview of promising sinomenine derivatives as potential RA drug therapies.


Asunto(s)
Artritis Reumatoide , Morfinanos , Neuralgia , Humanos , Artritis Reumatoide/tratamiento farmacológico , Morfinanos/uso terapéutico , Morfinanos/farmacología , Medicina Tradicional China , Neuralgia/tratamiento farmacológico
10.
J Control Release ; 348: 42-56, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35569587

RESUMEN

Sinomenine is a bioactive alkaloid isolated from the Chinese medicinal plant of Sinomenium acutum (Thunb.) Rehd.et Wils. Currently, sinomenine hydrochloride (SIN) preparations, classified as a natural disease-modifying anti-rheumatic drug (nDMARD), have been used for therapy of rheumatoid arthritis (RA); however, the efficacy of SIN was seriously limited by its short half-life, low bioavailability, and dose-dependent adverse reactions. In this study, a biomimetic nanocomplex based on Prussian blue nanoparticles (PB NPs) was developed for overcoming clinical limitations of SIN and accordingly improving its efficacy. In vitro studies showed that the nanocomplexes significantly inhibited abnormal proliferation of fibroblast-like synoviocytes (FLSs) by scavenging reactive oxygen species (ROS) and inhibiting secretion of proinflammatory cytokines. In vivo imaging demonstrated that the improved immune-escape properties of the nanocomplexes resulted in markedly increased half-life of circulation and levels of accumulated drugs at arthritic sites of adjuvant-induced arthritis (AIA) rats. Notably, the nanocomplexes significantly suppressed joint inflammation and protected against bone destruction of AIA rats by inhibiting inflammatory cytokine secretion of the synovial macrophages and FLSs. These results indicate that the nanocomplexes provide an excellent carrier for controlled release and targeted accumulation of SIN within the arthritic sites, which consequently achieve disease-remitting effects of SIN on RA.


Asunto(s)
Artritis Reumatoide , Morfinanos , Nanopartículas Multifuncionales , Animales , Artritis Reumatoide/tratamiento farmacológico , Citocinas , Morfinanos/farmacología , Morfinanos/uso terapéutico , Ratas
11.
J Oleo Sci ; 70(12): 1815-1828, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34866111

RESUMEN

Atherosclerosis (AS) is a cardiovascular disease that arise due to dysfunction of lipid deposition and metabolism. AS is causes the mortality and morbidity worldwide. Sinomenine isolated from the Sinomenium acutum is used extensively against the various cardiac diseases in China. However, the anti-atherosclerosis effect of sinomenine still not explore. In this study, we explore the cardioprotective and anti-atherosclerosis effect of sinomenine against Vitamin D3 and High fat induced atherosclerosis in rats. Sprague Dawley (SD) rats were used in this study. The rats were received the vitamin D (60000) and High fat diet to induce the atherosclerosis and divided into groups and received the oral administration of sinomenine (2.5, 5 and 10 mg/kg) and simvastatin (5 mg/kg). Body weight, organ weight and biochemical parameters were estimated. The mRNA expression of MyD88, TLR4, NF-κB and IκB were estimated. Sinomenine treated rats significantly (p<0.001) suppressed the body weight and modulated the organ weight (hepatic, renal and heart). Sinomenine significantly (p<0.001) decreased the level of triacylglycerols (TG), low density lipoprotein cholesterol (LDL-c), total cholesterol (TC), very low-density lipoprotein cholesterol (VLDL-c) and augmented the level of high-density lipoprotein cholesterol (HDL-c). Sinomenine treatment also reduced the level of atherogenic index (TC/HDL-c and LDL-c/HDL-c). Sinomenine treatment decrease the ratio of HMG CoA/Mevalonate and level of collagen and total protein. Sinomenine significantly (p<0.001) altered the level of heart parameters, antioxidant parameters and inflammatory cytokines. Sinomenine significantly (p<0.001) reduced the expression of MyD88, TLR4, NF-κB and IκB. Taken together, sinomenine exhibited the protective effect against the atherosclerosis via alteration of TLR4/NF-κB signaling pathway.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Aterosclerosis/etiología , Colecalciferol/efectos adversos , Dieta Alta en Grasa/efectos adversos , Morfinanos/administración & dosificación , Morfinanos/farmacología , Estrés Oxidativo/efectos de los fármacos , Fitoterapia , Administración Oral , Animales , Antioxidantes , Aterosclerosis/genética , Aterosclerosis/prevención & control , Citocinas/metabolismo , Inflamación , Mediadores de Inflamación/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Masculino , Morfinanos/aislamiento & purificación , FN-kappa B/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sinomenium/química , Receptor Toll-Like 4/metabolismo
12.
BMC Complement Med Ther ; 21(1): 284, 2021 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-34801005

RESUMEN

BACKGROUND: Stephania yunnanensis H. S. Lo is widely used as an antipyretic, analgesic and anti-inflammatory herbal medicine in SouthWest China. In this study, we investigated the anti-inflammatory activity and mechanism of sinoacutine (sino), one of the primary components extracted from this plant. METHODS: A RAW264.7 cell model was established using lipopolysaccharide (LPS) induced for estimation of cytokines in vitro, qPCR was used to estimate gene expression, western blot analysis was used to estimate protein level and investigate the regulation of NF- κB, JNK and MAPK signal pathway. In addition, an acute lung injury model was established to determine lung index and levels of influencing factors. RESULTS: Using the RAW264.7 model, we found that sino reduced levels of nitric oxide (NO), tumour necrosis factor-α (TNF-α), interleukin (IL)-1ß and prostaglandin E2 (PGE2) but increased levels of IL-6. qPCR analysis revealed that sino (50, 25 µg/ml) inhibited gene expression of nitric oxide synthase (iNOS). western blot analysis showed that sino significantly inhibited protein levels of both iNOS and COX-2. Further signalling pathway analysis validated that sino also inhibited phosphorylation of p65 in the NF-κB and c-Jun NH2 terminal kinase (JNK) signalling pathways but promoted the phosphorylation of extracellular signal regulated kinase (ERK) and p38 in the MAPK signalling pathway. In addition, in a mouse model induced by LPS, we determined that sino reduced the lung index and the levels of myeloperoxidase (MPO), NO, IL-6 and TNF-α in lung tissues and bronchoalveolar lavage fluid (BALF) in acute lung injury (ALI). CONCLUSION: Taken together, our results demonstrate that sino is a promising drug to alleviate LPS-induced inflammatory reactions.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Antiinflamatorios/farmacología , MAP Quinasa Quinasa 4/metabolismo , Morfinanos/farmacología , FN-kappa B/metabolismo , Extractos Vegetales/farmacología , Animales , Animales no Consanguíneos , Antiinflamatorios/química , China , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Estructura Molecular , Morfinanos/química , Extractos Vegetales/química , Células RAW 264.7
13.
Int J Nanomedicine ; 16: 3725-3739, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34103913

RESUMEN

PURPOSE: Transarterial chemoembolization is the preferred treatment for patients with middle and advanced-stage hepatocellular carcinoma (HCC); however, most hepatic artery embolization agents have various disadvantages. The purpose of this study was to evaluate phytantriol-based liquid crystal injections for potential use in treatment of HCC. METHODS: Using sinomenine (SN) and 5-fluorouracil (5-FU) as model drugs, three precursor in situ liquid crystal injections based on phytantriol (P1, P2, and P3) were prepared, and their in vitro biocompatibility, anticancer activity, and drug release investigated, to evaluate their feasibility for use in treatment of HCC. The properties of the precursor injections and subsequent cubic liquid crystal gels were observed by visual and polarizing microscopy, in an in vitro gelation experiment. Biocompatibility was evaluated by in vitro hemolysis, histocompatibility, and cytotoxicity assays. RESULTS: Precursor injections were colorless liquids that formed transparent cubic liquid crystal gels on addition of excess water. The three precursor injections all caused slight hemolysis, without agglutination, and were mildly cytotoxic. Histocompatibility experiments showed that P1 had good histocompatibility, while P2 and P3 resulted in strong inflammatory responses, which subsequently resolved spontaneously. In vitro anti-cancer testing showed that SN and 5-FU inhibited HepG2 cells in a time- and concentration-dependent manner and had synergistic effects. Further, in vitro release assays indicated that all three preparations had sustained release effects, with cumulative release of >80% within 48 h. CONCLUSION: These results indicate that SN and 5-FU have synergistic inhibitory effects on HepG2 cells, which has not previously been reported. Moreover, we describe a biocompatible precursor injection, useful as a drug carrier for the treatment of liver cancer, which can achieve targeting, sustained release, synergistic chemotherapy, and embolization. These data indicate that precursor injections containing SN and 5-FU have great potential for use in therapy for liver cancer.


Asunto(s)
Fluorouracilo/uso terapéutico , Cristales Líquidos/química , Neoplasias Hepáticas/tratamiento farmacológico , Morfinanos/uso terapéutico , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Muerte Celular , Portadores de Fármacos/química , Liberación de Fármacos , Sinergismo Farmacológico , Alcoholes Grasos/química , Fluorouracilo/farmacología , Geles , Hemólisis , Células Hep G2 , Humanos , Inyecciones , Morfinanos/farmacología , Ratas Sprague-Dawley , Agua/química
14.
Pharmacol Res ; 167: 105513, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33617975

RESUMEN

A large number of macrophages in inflamed sites not only amplify the severity of inflammatory responses but also contribute to the deleterious progression of many chronic inflammatory diseases, autoimmune diseases and cancers. Macrophage migration is a prerequisite for their entry into inflammatory sites and their participation of macrophages in the pathologic processes. Inhibition of macrophage migration is therefore a potential anti-inflammatory mechanism. Moreover, alleviation of inflammation also prevents the macrophages infiltration. Sinomenine (SIN) is an alkaloid derived from the Chinese medicinal plant Sinomenium acutum. It has multiple pharmacological effects, including anti-inflammation, immunosuppression, and anti-arthritis. However, its anti-inflammatory molecular mechanisms and effect on macrophage migration are not fully understood. The purpose of this research was to investigate the pharmacological effects and the molecular mechanism of SIN on macrophage migration in vivo and in vitro as well as to elucidate its anti-inflammatory mechanisms associated with macrophage migration. Our results showed that SIN reduced the number of RAW264.7 cells migrating into inflammatory paws and blocked lipopolysaccharide (LPS)-induced RAW264.7 cells and bone marrow-derived macrophages (BMDMs) migration in vitro. Furthermore, SIN attenuated the 3D mesenchymal migration of BMDMs. The absence of macrophage migration after circulatory and periphery macrophages depletion led to a reduction in the severity of inflammatory response. In macrophages depleted (macrophages-/-) mice, as inflammatory severity decreased, RAW264.7 cells migration was suppressed. A non-obvious effect of SIN on the inflammatory response was found in macrophages-/- mice, while the inhibitory effect of SIN on RAW264.7 cells migration was still observed. Furthermore, the migration of RAW264.7 cells pre-treated with SIN was suppressed in normal mice. Finally, Src/focal adhesion kinase (FAK)/P130Cas axis activation, which supports macrophages mesenchymal migration, and iNOS expression, NO production, integrin αV and in integrin ß3 expressions, which promote Src/FAK/P130Cas activation, were down-regulated by SIN. However, SIN had no obvious effect on the expression of the monocyte chemoattractant protein-1 (MCP-1), which is an important chemokine for macrophage migration. These results indicated that SIN significantly inhibited macrophage mesenchymal migration by down-regulating on Src/FAK/P130Cas axis activation. There was a mutual regulatory correlation between the inflammatory response and macrophage migration, and the effects of SIN on macrophage migration were involved in its anti-inflammatory activity.


Asunto(s)
Antiinflamatorios/farmacología , Movimiento Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Macrófagos/efectos de los fármacos , Morfinanos/farmacología , Animales , Antiinflamatorios/química , Proteína Sustrato Asociada a CrK/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Morfinanos/química , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Sinomenium/química , Familia-src Quinasas/metabolismo
15.
Bioorg Med Chem ; 31: 115986, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33412412

RESUMEN

Sinomenine is an alkaloid derived from Chinese medicinal plant Sinomenium acutum. Our previous studies suggested that sinomenine can inhibit the metastasis of breast cancer. However, whether sinomenine can inhibit the metastasis characteristics of breast cancer side population (SP) cells is still unknown. In present study, we isolated the side population (SP) cells from MDA-MB-231 cells by fluorescence-activated cell sorting (FACS). MDA-MB-231 SP cells were treated with different concentrations of sinomenine at the absence or presence of hypoxia, and cell viability were measured by CCK-8 assay. The transwell invasive assay were conducted to assess of the effect of sinomenine on the invasion of hypoxic MDA-MB-231 SP cells. The protein expression was detected by Western blot assay. Sinomenine inhibited the cell viability and invasion of hypoxic MDA-MB-231 SP cells. Western blot assay results showed that the upregulation of MMP-2 and MMP-9 by hypoxia was inversed by sinomenine. Additionally, it was found that sinomenine suppressed the activation of PI3K/Akt/mTOR pathway under hypoxia in MDA-MB-231 SP cells. Moreover, the inhibiton of sinomenine on metastasis of hypoxic MDA-MB-231SP cells and PI3K/Akt/mTOR pathway could be rescued by PI3K activator IGF-1. Our study suggested that sinomenine inhibits invasion of breast cancer SP cells under hypoxia through PI3K/Akt/mTOR pathway.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Hipoxia de la Célula/efectos de los fármacos , Morfinanos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Estructura Molecular , Proteínas Proto-Oncogénicas c-akt/metabolismo , Relación Estructura-Actividad , Serina-Treonina Quinasas TOR/metabolismo , Células Tumorales Cultivadas
16.
Cell Mol Neurobiol ; 41(5): 949-960, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32107752

RESUMEN

OBJECTIVE: Pharmacological evaluation of the mu-opioid receptor (MOR) agonist properties of NKTR-181 in rodent models. METHODS: Graded noxious stimulus intensities were used in rats to establish the antinociceptive potency and efficacy of NKTR-181 relative to morphine, fentanyl, and oxycodone. Characteristics of MOR agonist actions, as measured by antinociceptive tolerance and cross-tolerance, as well as opioid-induced hyperalgesia (OIH) and naloxone-precipitated withdrawal in NKTR-181- and morphine-dependent in mice, were compared. RESULTS: NKTR-181 showed dose- and time-related antinociception with similar maximal effects to morphine in the rat and mouse hot-water tail-flick test. No sex or species differences were observed in NKTR-181 or morphine antinociception. Rats treated with NKTR-181 and morphine exhibited decreases in both potency and maximal efficacy as nociceptive stimulus intensity was increased from a water temperature of 50 °C to 54 °C. Evaluation of antinociception at a high stimulus intensity revealed that oxycodone and fentanyl exhibited greater efficacy than either NKTR-181 or morphine. The relative potency difference between NKTR-181 and morphine across all tail-flick studies was determined to be 7.6-fold (90% confidence interval, 2.6, 21.5). The peak antinociceptive effect of NKTR-181 was delayed compared to that of the other opioids and cumulative drug effects were not observed. Repeated treatment with escalating, approximately equi-analgesic doses of NKTR-181 or morphine, produced antinociceptive tolerance and cross-tolerance. Under these pharmacological conditions, OIH and naloxone-precipitated physical dependence were similar for NKTR-181 and morphine. CONCLUSIONS: NKTR-181 had a slower onset, but similar efficacy, to morphine in the models studied supporting reduced abuse potential while maintaining analgesic effect in comparison with current opioids.


Asunto(s)
Analgésicos Opioides/farmacología , Morfinanos/farmacología , Morfina/farmacología , Dimensión del Dolor/efectos de los fármacos , Receptores Opioides mu/agonistas , Animales , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Femenino , Masculino , Ratones , Dimensión del Dolor/métodos , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/fisiología , Roedores
17.
J Mol Neurosci ; 71(2): 215-224, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32812186

RESUMEN

Multiple sclerosis (MS) is known as a chronic neuroinflammatory disorder typified by an immune-mediated demyelination process with ensuing axonal damage and loss. Sinomenine is a natural alkaloid with different therapeutic benefits, including anti-inflammatory and immunosuppressive activities. In this study, possible beneficial effects of sinomenine in an MOG-induced model of MS were determined. Sinomenine was given to MOG35-55-immunized C57BL/6 mice at doses of 25 or 100 mg/kg/day after onset of MS clinical signs till day 30 post-immunization. Analyzed data showed that sinomenine reduces severity of the clinical signs and to some extent decreases tissue level of pro-inflammatory cytokines IL-1ß, IL-6, IL-18, TNFα, IL-17A, and increases level of anti-inflammatory IL-10. In addition, sinomenine successfully attenuated tissue levels of inflammasome NLRP3, ASC, and caspase 1 besides its reduction of intensity of neuroinflammation, demyelination, and axonal damage and loss in lumbar spinal cord specimens. Furthermore, immunoreactivity for MBP decreased and increased for GFAP and Iba1 after MOG-immunization, which was in part reversed upon sinomenine administration. Overall, sinomenine decreases EAE severity, which is attributed to its alleviation of microglial and astrocytic mobilization, demyelination, and axonal damage along with its suppression of neuroinflammation, and its beneficial effect is also associated with its inhibitory effects on inflammasome and pyroptotic pathways; this may be of potential benefit for the primary progressive phenotype of MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Inflamasomas/antagonistas & inhibidores , Morfinanos/uso terapéutico , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Animales , Astrocitos/efectos de los fármacos , Peso Corporal , Citocinas/análisis , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Morfinanos/administración & dosificación , Morfinanos/farmacología , Glicoproteína Mielina-Oligodendrócito/inmunología , Glicoproteína Mielina-Oligodendrócito/toxicidad , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/toxicidad , Piroptosis/efectos de los fármacos , Distribución Aleatoria , Organismos Libres de Patógenos Específicos , Médula Espinal/química
18.
Mol Brain ; 13(1): 30, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-32127013

RESUMEN

Amyloid beta is a major constituent of the plaques found in the brains of patients suffering from Alzheimer's disease (AD). A growing body of research work suggests that neuroinflammation plays important roles in the development of AD. Thus, considerable efforts are directed towards identification of compounds that can reduce or inhibit neuroinflammation. Here, we show that sinomenine, a compound present in a Chinese medicinal plant, Sinomenium acutum, inhibits oligomeric amyloid beta-induced production of reactive oxygen species (ROS), nitric oxide (NO) and inflammation-related molecules from astrocytic cells. The conditioned medium from oligomeric amyloid beta-treated astrocytic cells induces cell death in the hippocampal neuronal cells. Importantly, sinomenine inhibits this cell death. In addition, this compound has inhibitory effects on the production of ROS, NO and inflammation-related factors from oligomeric amyloid-beta treated human astrocytes. Finally, the conditioned medium from oligomeric amyloid beta-treated human astrocytes induces cell death in the primary culture of human neurons, which is inhibited by sinomenine. Thus, sinomenine inhibits amyloid beta-induced production of toxic factors from astrocytes, and confers protection to hippocampal neuronal cells as well as human neurons against indirect toxicity. The results suggest that this compound could provide beneficial effects in AD and other neurodegenerative conditions by reducing inflammation and neuronal cell death.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Astrocitos/patología , Morfinanos/farmacología , Neuronas/patología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Línea Celular , Hipocampo/patología , Humanos , Inflamación/patología , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Óxido Nítrico/metabolismo , Especies Reactivas de Oxígeno/metabolismo
19.
J Integr Med ; 18(2): 174-180, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31992510

RESUMEN

OBJECTIVE: Premature and ill neonates undergo painful but medically necessary procedures while hospitalized. Although opiate drugs are administered as analgesics, problems associated with their side effects, tolerance, and potential dependence necessitate research into alternative pain-relieving medications. Here we test two plant-derived compounds in infant rats: sinomenine, which targets the Mas-related G-protein-coupled receptor member X2 opioid receptor; and salvinorin A, which is a κ opioid receptor agonist. In adult animals both sinomenine and salvinorin A are analgesic, but neither has been tested in infants. METHODS: We used the formalin and thermal plantar tests in rats 7 and 21 days of age (PN7 and PN21) for behavioral signs of pain. In addition, brain sections were stained using Fos immunohistochemistry to examine patterns of brain activation in the midbrain periaqueductal gray and the paraventricular nucleus of the hypothalamus. RESULTS: Sinomenine was analgesic in both the formalin and thermal tests on animals 21 days of age. At PN7 only the highest dose elevated response latencies in the thermal test and there were no effects of sinomenine in the formalin test. Analysis of Fos expression in the sinomenine-treated animals showed no drug effect, in contrast to the behavioral results. Salvinorin A was analgesic in the formalin test only at the highest dose at 21 days of age but not in the thermal test at either age. CONCLUSION: The increased modest effectiveness of sinomenine in older animals and the minimum salvinorin A drug effect suggest that the compounds act on sites that develop during the preweaning period (sinomenine) or after weaning (salvinorin A).


Asunto(s)
Analgésicos/uso terapéutico , Diterpenos de Tipo Clerodano/uso terapéutico , Morfinanos/uso terapéutico , Dolor/tratamiento farmacológico , Receptores Opioides/agonistas , Salvia/química , Sinomenium/química , Analgésicos/farmacología , Analgésicos Opioides/farmacología , Analgésicos Opioides/uso terapéutico , Animales , Conducta Animal , Diterpenos de Tipo Clerodano/farmacología , Calor , Humanos , Lactante , Recién Nacido , Morfinanos/farmacología , Dimensión del Dolor , Fitoterapia , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Ratas Long-Evans , Receptores Opioides kappa/agonistas
20.
J Vet Pharmacol Ther ; 42(6): 732-737, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31490576

RESUMEN

The aim of the research was to investigate the anti-endotoxin and anti-inflammatory effects of Sinomenine, an agent commonly found in Chinese herbal medicines. Endotoxin (i.e., 1 mg lipopolysaccharide (LPS)/kg)) was administered via intraperitoneal (IP) injection to piglets in high-, middle-, and low-dose sinomenine groups. Piglets were then treated with 1, 5 or 10 mg/kg sinomenine, intramuscularly (i.m.), 3 hr after LPS. Vehicle was administered, as above, to drug control group piglets followed 3 hr later by 10 mg/kg sinomenine i.m.. LPS control group piglets were challenged with 1 mg/kg LPS IP, followed by vehicle i.m., and naïve control piglets were treated with normal saline IP, followed by normal saline i.m., as above. Temperatures were measured, and blood samples were collected from the precaval veins of piglets at 12, 24, and 48 hr post-LPS or vehicle injection. Clinical signs were recorded, and index levels were analyzed via ELISA. Sinomenine was found to reduce the incidence and severity of LPS-induced toxicities, including body temperature elevation, cell adhesion, and systemic inflammation. These data suggest that sinomenine may be effective for regulating inflammatory responses and has the potential for use as an anti-endotoxin therapy.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Proteínas Portadoras/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Lipopolisacáridos/toxicidad , Antígeno de Macrófago-1/metabolismo , Glicoproteínas de Membrana/metabolismo , Morfinanos/farmacología , Porcinos , Proteínas de Fase Aguda/genética , Animales , Proteínas Portadoras/genética , Relación Dosis-Respuesta a Droga , Selectina L , Antígeno de Macrófago-1/genética , Masculino , Glicoproteínas de Membrana/genética , Morfinanos/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA