Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Pharm Pharmacol ; 76(4): 426-434, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38290061

RESUMEN

OBJECTIVES: Sanshimao (SSM) is a traditional Chinese medicine formula for advanced hepatocellular carcinoma (HCC). This study was designed to investigate the effect of SSM on HCC-induced angiogenesis and to explore the potential mechanism. METHODS: The endothelial cells were cultured with HCC cells conditioned medium in the 1% oxygen atmosphere to imitate tumor hypoxia microenvironment. EA.hy926 cells migration and tubulogenesis were detected by tube formation and scratch-wound assay. The protein microarray was employed to explore SSM-targeted proteins in Huh7 cells. We also established an animal model to observe the effects of SSM on angiogenesis in vivo. RESULTS: The data indicated that SSM reduced HCC-induced migration and tube formation of EA.hy926 cells at low dose under hypoxic conditions. These effects might be partly owing to suppression of HIF-1α-induced vascular endothelial growth factorα expression in Huh7 cells. Moreover, this inhibition was in an MKK6/P38-dependent way. Besides, Huh7 subcutaneous tumor models in nude mice further demonstrated the inhibition of SSM on tumor weight might be exerted partly by reduction of angiogenesis via blocking MKK6/P38 signaling pathways. CONCLUSION: SSM inhibits HCC-induced pro-angiogenesis under hypoxic conditions via suppression of MKK6/P38 signaling pathways, which is favorable for HCC tumor growth.


Asunto(s)
Carcinoma Hepatocelular , Medicamentos Herbarios Chinos , Neoplasias Hepáticas , Neovascularización Patológica , Animales , Ratones , Angiogénesis , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Células Endoteliales/metabolismo , Hipoxia/tratamiento farmacológico , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Transducción de Señal , Microambiente Tumoral , Medicamentos Herbarios Chinos/farmacología , MAP Quinasa Quinasa 6/efectos de los fármacos , MAP Quinasa Quinasa 6/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
2.
Nutrition ; 118: 112273, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38096603

RESUMEN

BACKGROUND: Skeletal muscle synthesizes, stores, and releases body L-glutamine (GLN). Muscle atrophy due to disabling diseases triggers the activation of proteolytic and pro-apoptotic cell signaling, thus impairing the body's capacity to manage GLN content. This situation has a poor therapeutic prognosis. OBJECTIVE: Evaluating if oral GLN supplementation can attenuate muscle wasting mediated by elevated plasma cortisol and activation of caspase-3, p38MAPK, and FOXO3a signaling pathways in soleus and gastrocnemius muscles of rats submitted to 14-day bilateral hindlimbs immobilization. METHODS: Animals were randomly distributed into six groups: non-immobilized rats (Control), control orally supplemented with GLN (1 g kg-1) in solution with L-alanine (ALA: 0.61 g kg-1; GLN+ALA), control orally supplemented with dipeptide L-alanyl-L-glutamine (DIP; 1.49 g kg-1), hindlimbs immobilized rats (IMOB), IMOB orally GLN+ALA supplemented (GLN+ALA-IMOB), and IMOB orally DIP supplemented (DIP-IMOB). Plasma and muscle GLN concentration, plasma cortisol level, muscle caspase-3 activity, muscle p38MAPK and FOXO3a protein content (total and phosphorylated forms), and muscle cross-sectional area (CSA) were measured. RESULTS: Compared to controls, IMOB rats presented: a) increased plasma cortisol levels; b) decreased plasma and muscle GLN concentration; c) increased muscle caspase-3 activity; d) increased total and phosphorylated p38MAPK protein content; e) increased FOXO3a and decreased phosphorylated FOXO3a protein content; f) reduced muscle weight and CSA befitting to atrophy. Oral supplementation with GLN+ALA and DIP was able to significantly attenuate these effects. CONCLUSIONS: These findings attest that oral GLN supplementation in GLN+ALA solution or DIP forms attenuates rats' skeletal muscle mass wasting caused by disuse-mediated muscle atrophy.


Asunto(s)
Glutamina , Hidrocortisona , Atrofia Muscular , Animales , Ratas , Caspasa 3/metabolismo , Suplementos Dietéticos , Dipéptidos/metabolismo , Dipéptidos/farmacología , Dipéptidos/uso terapéutico , Glutamina/farmacología , Músculo Esquelético , Atrofia Muscular/tratamiento farmacológico , Atrofia Muscular/etiología , Atrofia Muscular/metabolismo , Transducción de Señal , Proteína Forkhead Box O3/efectos de los fármacos , Proteína Forkhead Box O3/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
3.
Int J Mol Sci ; 23(3)2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-35163281

RESUMEN

Calycosin, a bioactive isoflavonoid isolated from root extracts of Astragalus membranaceus, has been reported to inhibit melanogenesis, the mechanism of which remains undefined. In this study, we interrogated the mechanistic basis by which calycosin inhibits melanin production in two model systems, i.e., B16F10 melanoma cells and zebrafish embryos. Calycosin was effective in protecting B16F10 cells from α-melanocyte-stimulating hormone (α-MSH)-induced melanogenesis and tyrosinase activity. This anti-melanogenic effect was accompanied by decreased expression levels of microphthalmia-associated transcription factor (MITF), a key protein controlling melanin synthesis, and its target genes tyrosinase and tyrosinase-related protein-2 (TRP-2) in calycosin-treated cells. Mechanistically, we obtained the first evidence that calycosin-mediated MITF downregulation was attributable to its ability to block signaling pathways mediated by cAMP response element-binding protein (CREB) and p38 MAP kinase. The protein kinase A (PKA) inhibitor H-89 and p38 inhibitor SB203580 validated the premise that calycosin inhibits melanin synthesis and tyrosinase activity by regulating the PKA/CREB and p38 MAPK signaling pathways. Moreover, the in vivo anti-melanogenic efficacy of calycosin was manifested by its ability to suppress body pigmentation and tyrosinase activity in zebrafish embryos. Together, these data suggested the translational potential of calycosin to be developed as skin-lightening cosmeceuticals.


Asunto(s)
Isoflavonas/farmacología , Melaninas/metabolismo , Animales , Astragalus propinquus/metabolismo , Línea Celular Tumoral , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Isoflavonas/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo , Fosforilación/efectos de los fármacos , Extractos Vegetales/farmacología , Raíces de Plantas , Transducción de Señal/efectos de los fármacos , Pez Cebra/metabolismo , alfa-MSH/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
4.
Pharmacology ; 106(11-12): 623-636, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34753130

RESUMEN

INTRODUCTION: 5-Fluorouracil (5-FU) is used to treat various cancers, including non-small-cell lung cancer (NSCLC). It inhibits nucleotide synthesis and induces single- and double-strand DNA breaks. In the homologous recombination pathway, radiation-sensitive 52 (Rad52) plays a crucial role in DNA repair by promoting the annealing of complementary single-stranded DNA and stimulating Rad51 recombinase activity. Erlotinib (Tarceva) is a selective epidermal growth factor receptor tyrosine kinase inhibitor with clinical activity against NSCLC cells. However, whether the combination of 5-FU and erlotinib has synergistic activity against NSCLC cells is unknown. METHODS: After the 5-FU and/or erlotinib treatment, the expressions of Rad52 mRNA were determined by quantitative real-time polymerase chain reaction analysis. Protein levels of Rad52 and phospho-p38 MAPK were determined by Western blot analysis. We used specific Rad52 or p38 MAPK small interfering RNA and p38 MAPK inhibitor (SB2023580) to examine the role of p38 MAPK-Rad52 signal in regulating the chemosensitivity of 5-FU and/or erlotinib. Cell viability was assessed by MTS assay and trypan blue exclusion assay. RESULTS: In 2 squamous cell carcinoma cell lines, namely, H520 and H1703, 5-FU reduced Rad52 expression in a p38 MAPK inactivation-dependent manner. Enhancement of p38 MAPK activity by transfection with MKK6E (a constitutively active form of MKK6) vector increased the Rad52 protein level and cell survival by 5-FU. However, in human lung bronchioloalveolar cell adenocarcinoma A549 cells, 5-FU reduced Rad52 expression and induced cytotoxicity independent of p38 MAPK. Moreover, 5-FU synergistically enhanced the cytotoxicity and cell growth inhibition of erlotinib in NSCLC cells; these effects were associated with Rad52 downregulation and p38 MAPK inactivation in H520 and H1703 cells. CONCLUSION: The results provide a rationale for combining 5-FU and erlotinib in lung cancer treatment.


Asunto(s)
Antineoplásicos/farmacología , Clorhidrato de Erlotinib/farmacología , Fluorouracilo/farmacología , Neoplasias Pulmonares/patología , Neoplasias de Células Escamosas/patología , Proteína Recombinante y Reparadora de ADN Rad52/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Humanos
5.
Pak J Pharm Sci ; 34(2): 585-589, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34275833

RESUMEN

This study aims to explore the mechanism of NSAID-related gastric ulcer treated by JIA WEI WU QI SAN. Clean-grade SD rats were randomly divided into four groups. Group A was assigned as the control group. Groups B, C and D were intragastrically administered with 2.5mg/kg of indomethacin solution QD after 48 hours. After 15 days of treatment, group B was administered with 0.9% sodium chloride, group C was given rabeprazole (2mg/kg), and group D was administered with JIA WEI WU QI SAN (2g/kg). Abdominal aorta sampling was performed, and gastric tissues were isolated on the 29th day. The protein expression of p-P38MAPK and COX-2 were detected by western blot, while the concentration of PGE2 and IL-1 were determined by ELISA. (1) The expression of IL-1ingroup B dramatically declined in group D (P<0.01). (2)The expression of PGE-2dramatically increased in group D(P<0.01). (3) The expression of COX-2 increased in group D (P<0.05). (4) The expression of p-P38MAPK decreased in group D (P<0.05). JIA WEI WU QI SAN has multiple functions, including the activation of the p-P38MAPK signaling pathway, which promote the activation of COX-2, induce the arachidonic acid to increase the level of PG, and decrease the concentration of IL-1, thereby inducing an inflammatory reaction, and promote gastric mucosa repair.


Asunto(s)
Antiinflamatorios no Esteroideos/efectos adversos , Dinoprostona/metabolismo , Medicamentos Herbarios Chinos/farmacología , Interleucina-1/metabolismo , Úlcera Gástrica/metabolismo , Estómago/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Animales , Ácido Araquidónico/metabolismo , Ciclooxigenasa 2/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Indometacina/efectos adversos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Estómago/metabolismo , Úlcera Gástrica/inducido químicamente , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
J BUON ; 25(1): 314-318, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32277648

RESUMEN

PURPOSE: Gastric cancer accounts for considerable mortality across the globe. In this study the anticancer effects of a natural compound Berberine were investigated in vitro. Effects of berberine on cell migration, cellular apoptosis, Nf-kB and JNK/p38 signalling pathways were also studied. METHODS: The cell viability of SNU-1 gastric cancer cells after berberine treatment was evaluated by CCK-8 assay, while the effects on cell migration were checked by wound healing assay. Effects on cellular apoptosis were evaluated by fluorescence microscopy using DAPI staining, as well as using flow cytometry with annexin V/propidium iodide (PI) staining. Effects on apoptosis-related protein expressions were checked by western blot method. RESULTS: The results showed that Berberine decreased the viability of the gastric cancer SNU-1 cells and exhibited an IC50 of 30 µM. The cytoxicity of Berberine was also investigated on the normal GES-1 gastric cells and it was found that Berberine exerted very low toxic effects on these cells and exhibited an IC50 of 120 µM. Berberine also caused remarkable changes in the morphology of the SNU-1 cells. PI and DAPI staining revealed that Berberine prompted apoptosis of the SNU-1 cells in a dose-dependent manner. The apoptotic cells increased from 2.2% in control to around 35% at 30 µM concentration. Berberine also suppressed the migration and invasion of the gastric cancer cells via blocking of the JNK/p38 signalling pathway. CONCLUSIONS: Berberine may act as a promising drug candidate for gastric cancer as demonstrated from the current study.


Asunto(s)
Apoptosis/efectos de los fármacos , Berberina/uso terapéutico , FN-kappa B/antagonistas & inhibidores , Neoplasias Gástricas/tratamiento farmacológico , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Berberina/farmacología , Humanos , Neoplasias Gástricas/patología
7.
Med Sci Monit ; 25: 7538-7546, 2019 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-31590176

RESUMEN

BACKGROUND Lung cancer is the leading cause of cancer deaths in the world. Its major histopathological subtype is non-small cell lung cancer (NSCLC). Xiaoai Jiedu recipe (XJR) is a traditional Chinese medicine formula that can suppress growth and invasion of tumor cells. Here, we assessed the antitumor effect of XJR on NSCLC explored the underlying mechanisms. MATERIAL AND METHODS Three concentrations of XJR (low, middle, and high) were used to treat A549 cells. Cell Counting Kit-8 and colony formation assay were used to measure proliferation of A549 cells. Apoptosis was evaluated by Hoechst 33342 staining and flow cytometry. The expression of apoptosis-associated proteins was measured by Western blot analysis. Transwell and scratch wound healing assay were used to assess invasion and migration, respectively, of A549 cells. The expression of p38 MAPK pathway-associated proteins were measured using Western blot analysis. RESULTS XJR suppressed proliferation and promoted apoptosis of A549 cells, especially in the high-dose group. The expression of Bcl-2 was reduced with increasing expression of Bax, cleaved caspase-3, and cleaved caspase-9. Invasion and migration abilities of A549 cells were inhibited after XJR treatment. XJR treatment decreased the expression levels of phosphorylated p38 (p-p38), p-ERK, and p-JNK in a dose-dependent manner. CONCLUSIONS The results demonstrated that XJR can inhibit proliferation, invasion, and migration, and induce apoptosis of NSCLC by blocking the p38 MAPK pathway, which shows the potential of XJR as a new treatment of NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Medicamentos Herbarios Chinos/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Células A549 , Animales , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , China , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Medicina Tradicional China/métodos , Invasividad Neoplásica/fisiopatología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Artículo en Chino | MEDLINE | ID: mdl-31245955

RESUMEN

OBJECTIVE: To observe the effects of Shenmai injection(SM) on p38MAPK and the apoptosis-related genes in lung injury induced by intestinal ischemia reperfusion (I/R) in rats and to investigate the protective mechanism of SM. METHODS: Rat model of intestinal I/R injury was established with clamping of the superior mesenteric artery (SMA) for 60 min and then clamping was relieved for 60 min. Twenty-four SD rats were randomly divided into three groups with eight rats in each: control group, intestinal ischemia/reperfusion group(I/R group), Shenmai injection treated group (SM+I/R group). Lung wet/dry weight ratio(W/D), the contents of phosphatidylcholine (PC) and total phospholipid(TPL) which are the major ingredients of pulmonary surfactant were measured, as well as the expression levels of p38MAPK, Bcl-2 and Bax proteins in lung tissue were examined by using immunohistochemical method. RESULTS: Compared with control group, lung W/D was significantly increased, the contents of PC and TPL were significantly decreased, the protein expression levels of p38MAPK, Bcl-2 and Bax were significantly increased in I/R group (all P<0.01). But Bax protein expression was much greater than Bcl-2 protein expression, the ratio of Bcl-2 to Bax were significantly decreased in I/R group than that in control group (P<0.01). Compared with I/R group, lung W/D was significantly decreased, while the contents of PC and TPL were significantly increased, the p38MAPK and Bax protein expression levels were significantly decreased in SM+I/R group (all P<0.01); both Bcl-2 protein expression and the ratio of Bcl-2 to Bax were significantly increased in SM+I/R group than those in I/R group (P<0.01). The correlation analysis indicated that the expression level of p38MAPK protein in lung tissue was negatively correlated with the contents of PC and the ratio of Bcl-2 to Bax (r is -0.787 and -0.731, all P<0.01). CONCLUSION: SM can protect the lung injury induced by intestinal I/R injury, which may be mediated by inhibiting the activation of p38MAPK, improving the ratio of Bcl-2 to Bax to inhibit lung apoptosis.


Asunto(s)
Medicamentos Herbarios Chinos , Lesión Pulmonar , Daño por Reperfusión , Proteínas Quinasas p38 Activadas por Mitógenos , Animales , Apoptosis , Combinación de Medicamentos , Medicamentos Herbarios Chinos/farmacología , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/genética , Proteínas Proto-Oncogénicas c-bcl-2 , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/complicaciones , Proteína X Asociada a bcl-2 , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Phytomedicine ; 58: 152864, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30878874

RESUMEN

BACKGROUND: Ulcerative colitis (UC) is a chronic inflammatory condition of the intestines and is difficult to cure once diagnosed. The efficacy of the current clinical treatment for UC is limited. Common anti-inflammatory drugs are prone to adverse effects, while novel biological agents are expensive, although tolerated by patients. Therefore, an urgency exists to find more safe and effective drugs to treat UC. Osthole is an active constituent isolated from the fruit of Cnidium monnieri (L.) Cuss. Osthole has anti-inflammatory activities and offers certain intestinal protection. These characteristics indicate that osthole has the potential to inhibit UC. PURPOSE: The study was conducted to investigate the anti-inflammatory potential of osthole in LPS-induced RAW 264.7 cells and dextran sulphate sodium (DSS)-induced ulcerative colitis in mice. METHODS: In in vitro experiments, mouse monocyte-macrophage RAW 264.7 cells were stimulated by 1 µg/ml LPS to produce inflammatory mediators. Griess reagent was used to determine Nitric Oxide (NO) production, and ELISA kits were used to determine the levels of PGE2, TNF-α, and IL-6. The anti-inflammatory mechanisms of osthole were detected using western blot. In in vivo experiments, UC was induced via the intragastric administration of 3.5% DSS to BALB/C mice for 7 days. During the experiment, clinical signs and body weight were monitored and recorded daily to calculate the DAI score. At the end of the experiment, the colon lengths were measured. The colonic histopathological lesions were evaluated. MPO activity and TNF-α levels were determined using the corresponding kits. The protein expression of TNF-α and NF-κB pathways were analysed using western blot. RESULTS: In an in vitro study, osthole inhibited the production of NO, PGE2, TNF-α, and IL-6 in LPS-induced RAW 264.7 cells. The results of western blot showed that osthole inhibited the expression of iNOS, COX-2, p38 MAPK and IκB α in RAW 264.7 cells. On this basis, in DSS-induced UC mice, it was found that osthole relieved the symptoms of UC by inhibiting weight loss, colon shortening and the DAI score, and simultaneously alleviating colon tissue lesions. It was also found that osthole reduced the levels of TNF-α in serum and colon tissues and effectively inhibited the activity of MPO. The western blot results showed that osthole reduced the expression of NF-κB p65 and p-IκB α and increased the content of IκB α in colon tissues. CONCLUSION: Osthole exerted anti-inflammatory effects by blocking the activation of the NF-κB and MAPK/p38 pathways. Additionally, osthole possesses therapeutic potential in the treatment of UC.


Asunto(s)
Antiinflamatorios/farmacología , Cnidium/química , Colitis Ulcerosa/tratamiento farmacológico , Cumarinas/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Colitis Ulcerosa/patología , Dinoprostona/metabolismo , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos BALB C , Inhibidor NF-kappaB alfa/metabolismo , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Células RAW 264.7 , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Med Sci Monit ; 25: 2002-2008, 2019 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-30879017

RESUMEN

BACKGROUND Murrayanine is a carbazole alkaloid derived from Murraya koenigii, which has been used in traditional Chinese medicine in the treatment of cancer. This study aimed to investigate the effects of murrayanine on human lung adenocarcinoma cells in vitro and to investigate the mechanisms of its action. MATERIAL AND METHODS A549 human lung adenocarcinoma cells and MRC-5 human lung fibroblasts were grown in culture, and an MTT assay determined cell viability. Cells were treated for 24 h with increasing doses of murrayanine (0, 9, 18, and 36 µM). Fluorescence, using 4', 6-diamidino-2-phenylindole (DAPI), acridine orange, ethidium bromide, and propidium iodide (PI), were used for the detection of apoptosis. The cell cycle was studied with fluorescence-activated cell sorting (FACS), and Western blot evaluated protein expression. RESULTS Murrayanine treatment resulted in significant dose-dependent inhibition of the growth of A549 cells (p<0.05), with an IC50 of 9 µM, and arrested the cells at the G2/M phase of the cell cycle, reduced the expression of cyclin D and E, CDK2, 4, and 6, and increased the expression of p21 and p27. Murrayanine treatment increased apoptosis of the A549 cells and increased cleaved of caspase-3 and caspase-9, and the Bax/Bcl-2 ratio. Murrayanine treatment increased levels of reactive oxygen species (ROS), disrupted the mitochondrial membrane potential, inhibited invasion, and inhibited phosphorylation of p38 mitogen-activated protein kinase (MAPK) of the A549 cells. CONCLUSIONS Murrayanine induced cell cycle arrest, oxidative stress, and inhibited the expression of phosphorylated p38 in A549 adenocarcinoma cells.


Asunto(s)
Adenocarcinoma del Pulmón/tratamiento farmacológico , Carbazoles/farmacología , Células A549 , Adenocarcinoma/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Carbazoles/metabolismo , Caspasa 3/efectos de los fármacos , Caspasa 9/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , China , Fase G2/efectos de los fármacos , Humanos , Neoplasias Pulmonares/patología , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos
11.
Neuroreport ; 30(6): 434-440, 2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30817685

RESUMEN

Ginkgo biloba extract EGb 761 possesses a variety of biological effects and has been proved to be beneficial in Alzheimer's disease. This study aimed to explore the anti-inflammatory mechanisms of EGb 761 on the Aß1-42-induced BV-2 microglial cells. We analyzed the production and gene expression of proinflammatory cytokines by enzyme-linked immunosorbent assay and qRT-PCR, examined phosphorylation of MAPKs by western blot and measured nuclear factor-κB nuclear translocation. Compared with Aß1-42-treated group, EGb 761 inhibited release and gene expression of tumor necrosis factor-α and interleukin-1ß, suppressed nuclear translocation of nuclear factor-κB and attenuated phosphorylation of p38 MAPK in a concentration-dependent manner, but not ERK and JNK. In summary, the results suggested that EGb 761 could attenuate Aß1-42-induced neuroinflammatory response.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Inflamación/metabolismo , Microglía/efectos de los fármacos , Extractos Vegetales/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Animales , Línea Celular , Ginkgo biloba , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Microglía/metabolismo
12.
Biomed Pharmacother ; 108: 1081-1089, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30372808

RESUMEN

BACKGROUND: Tongmai Yangxin Pills (TMYXP), a traditional Chinese patent medicine, has been widely used to treat coronary heart disease for few decades. However, the potential protective effect of TMYXP on cisplatin-induced cardiotoxicity has not been reported. METHODS: The target proteins corresponding to compounds from Pharmmapper database, PubMed database and ChEMBL database were collected and construct a 'TMYXP-compound-target' network. DAVID database was used for annotation and enrichment pathways and String 9.1 database was used for analysis the protein-protein interaction. Cisplatin-induced rat cardiotoxicity model was established to verify the protective effects mechanism of TMYXP. RESULTS: The target proteins corresponding to compounds from multiple databases were collected and construct a TCM-compound-target network to enriched pathways with high enrichment score. GO analysis and enrichment clusters point that response of oxidative stress is the main biological process of TMYXP, and Nrf2 signaling pathway and MAPK signaling pathway might be the key functional pathways. In vivo experiments, we proved that TMYXP improves anti-oxidative stress ability and reduce apoptosis through regulating Nrf2/HO-1 pathway and p38-MAPK pathway. CONCLUSION: The effects of TMYXP on regulate cardiomyocyte free radical balance and reduce apoptosis, making it possible as a drug candidate for platinum chemotherapeutic induced cardiac injury.


Asunto(s)
Antioxidantes/uso terapéutico , Cardiotoxicidad/tratamiento farmacológico , Cardiotoxicidad/patología , Cisplatino/efectos adversos , Medicamentos Herbarios Chinos/uso terapéutico , Estrés Oxidativo , Mapas de Interacción de Proteínas , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Cardiotoxicidad/fisiopatología , Núcleo Celular/metabolismo , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/farmacología , Corazón/efectos de los fármacos , Corazón/fisiopatología , Masculino , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos , Mapas de Interacción de Proteínas/efectos de los fármacos , Ratas Wistar , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos
13.
Endocrinology ; 159(9): 3351-3364, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30010822

RESUMEN

Estrogen receptor α (ERα) is a ligand-activated transcriptional activator that is also involved vascular inflammation and atherosclerosis. Whether different ligands may affect this activity has not been explored. We screened a panel of phytoestrogens for their role in ERα binding and transcriptional transcription, and correlated the findings to anti-inflammatory activities in vascular endothelial cells stably expressing either a wild-type or mutant form of ERα deficient in its membrane association. Taxifolin and silymarin were "high binders" for ERα ligand binding; quercetin and curcumin were "high activators" for ERα transactivation. Using these phytoestrogens as functional probes, we found, in endothelial cells expressing wild-type ERα, the ERα high activator, but not the ERα high binder, promoted ERα nuclear translocation, estrogen response element (ERE) reporter activity, and the downstream gene expression. In endothelial cells expressing membrane association-deficient mutant ERα, the ERα nuclear translocation was significantly enhanced by taxifolin and silymarin, which still failed to activate ERα. Inflammation response was examined using the systemic or vascular inflammation inducers lipopolysaccharide or oxidized low-density lipoprotein. In both cases, only the ERα high activator inhibited nuclear translocation of nuclear factor κB, JNK, and p38, and the production of inflammatory cytokines IL-1ß and TNFα. We confirm a threshold nuclear accumulation of ERα is necessary for its transactivation. The anti-inflammatory activity of phytoestrogens is highly dependent on ERα transactivation, less so on the ligand binding, and independent of its membrane association. A pre-examination of phytoestrogens for their mode of ERα interaction could facilitate their development as better targeted receptor modifiers.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Antioxidantes/farmacología , Células Endoteliales/efectos de los fármacos , Receptor alfa de Estrógeno/efectos de los fármacos , Fitoestrógenos/farmacología , Aterosclerosis/inmunología , Línea Celular , Curcumina/farmacología , Células Endoteliales/metabolismo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Humanos , Inflamación/inmunología , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/inmunología , Ligandos , Lipopolisacáridos/farmacología , Lipoproteínas LDL/farmacología , MAP Quinasa Quinasa 4/efectos de los fármacos , MAP Quinasa Quinasa 4/inmunología , Simulación del Acoplamiento Molecular , Mutación , FN-kappa B/efectos de los fármacos , FN-kappa B/inmunología , Transporte de Proteínas , Quercetina/análogos & derivados , Quercetina/farmacología , Elementos de Respuesta , Transducción de Señal , Silimarina/farmacología , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
14.
Rev Neurosci ; 30(1): 9-30, 2018 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29804103

RESUMEN

A myriad of environmental and genetic factors, as well as the physiologic process of aging, contribute to Alzheimer's disease (AD) pathology. Neuroinflammation is and has been a focus of interest, as a common gateway for initiation of many of the underlying pathologies of AD. Amyloid beta (Aß) toxicity, increasing RAGE expression, tau hyperphosphorylation, induction of apoptosis, and deregulated autophagy are among other mechanisms, partly entangled and being explained by activation of mitogen-activated protein kinase (MAPK) and MAPK signaling. p38 MAPK is the most essential regulator of Aß induced toxicity from this family. p38 induces NF-κB activation, glutamate excitotoxicity, and disruption of synaptic plasticity, which are other implications of all justifying the p38 MAPK as a potential target to break the vicious Aß toxicity cycle. Until recently, many in vivo and in vitro studies have investigated the effects of p38 MAPK inhibitors in AD. The pyridinyl imidazole compounds SB202190 and SB203580 have shown promising anti-apoptotic results in vivo. MW108 inhibits activation of p38 and is able to postpone cognitive decline in animal models. The PD169316, with anti-inflammatory, anti-oxidative, and anti-apoptotic features, has improved spatial memory in vivo. Natural compounds from Camellia sinensis (green tea), polyphenols from olive oil, pinocembrin from propolis, and the puerarine extract isoflavones, have shown strong anti-apoptotic features, mediated by p38 MAPK inhibition. Use of these drug targets is limited due to central nervous system side effects or cross-reactivity with other kinases, predicting the low efficacy of these drugs in clinical trials.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/efectos adversos , Inhibidores Enzimáticos/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Humanos , Receptor para Productos Finales de Glicación Avanzada/metabolismo
15.
Int J Mol Sci ; 18(7)2017 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-28661460

RESUMEN

Dysregulated human monocytes/macrophages can synthesize and secrete matrix metalloproteinases (MMPs), which play important roles in the progression of sepsis. In this study, we investigated the effects and mechanism of a novel histone deacetylase (HDAC8) inhibitor, (E)-N-hydroxy-4-methoxy-2-(biphenyl-4-yl)cinnamide (WK2-16), on MMP-9 production and activation in stimulated human monocytic THP-1 cells. Our results demonstrated that the acetylation level of structural maintenance of chromosomes 3 (SMC3) was up-regulated by WK2-16 in THP-1 cells. Consistently, an in vitro enzyme study demonstrated that WK2-16 selectively inhibited HDAC8 activity. Moreover, the WK2-16 concentration dependently suppressed MMP-9-mediated gelatinolysis induced by tumor necrosis factor-α (TNF-α) or lipopolysaccharide (LPS). Additionally, WK2-16 significantly inhibited both MMP-9 protein and mRNA expression without cellular toxicity. Nevertheless, WK2-16 suppressed the extracellular levels of interleukin (IL)-6 from LPS-stimulated THP-1 cells. For the signaling studies, WK2-16 had no effect on LPS/TLR4 downstream signaling pathways, such as the NF-κB and ERK/JNK/P38 MAPK pathways. On the other hand, WK2-16 enhanced the recruitment of acetylated Yin Yang 1 (YY1) with HDAC1. Finally, in vivo studies indicated that WK2-16 could reduce the serum levels of TNF-α and IL-6 in endotoxemic mice. These results suggested that HDAC8 inhibition might provide a novel therapeutic strategy of hypercytokinemia in sepsis.


Asunto(s)
Citocinas/efectos de los fármacos , Citocinas/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/efectos de los fármacos , Histona Desacetilasas/metabolismo , Lipopolisacáridos/farmacología , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas Represoras/efectos de los fármacos , Proteínas Represoras/metabolismo , Acetilación , Animales , Proteínas de Ciclo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Ciclooxigenasa 2/efectos de los fármacos , Regulación hacia Abajo , Endotoxemia , Histona Desacetilasa 1/efectos de los fármacos , Humanos , Interleucina-6 , Proteínas Quinasas JNK Activadas por Mitógenos/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/metabolismo , FN-kappa B/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/efectos de los fármacos , Sepsis/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Células THP-1/efectos de los fármacos , Tubulina (Proteína)/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Factor de Transcripción YY1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos
16.
Microcirculation ; 24(7)2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28597598

RESUMEN

OBJECTIVE: Although Rosa rugosa has been applied for preventing coronary artery disease, the pharmacological mechanism is little explored. In this study, the effects and mechanisms of Rosa rugosa flavonoids (RRF) on myocardial ischemia reperfusion injury (MIRI) were investigated. METHODS: Mice were pretreated by intragastric administration of 600 mg/kg RRF for 7 days. Then MIRI was induced by 45 minutes coronary artery ligation and 3 hours reperfusion. Myocardial infarct size (MIS) and histopathology, activities of myocardial enzymes, and effects of RRF on inflammation and apoptosis were evaluated. RESULTS: Pretreating the mice with RRF significantly reduced MIS and inhibited activity of plasma myocardial enzymes. Activity of the enzymes associated with anti-oxidation, SOD, and TEAC, and mRNA expression of NOX2 were significantly elevated. RRF pretreatment significantly decreased the translocation of p65 from the cytoplasm into the nucleus and reduced the expression of the pro-inflammatory cytokines, IL-6 and IL-1ß. RRF pretreatment also significantly prevented the expression of caspase-3 and Bax, and increased the expression of Bcl-2. And RRF inhibited the phosphorylation of JNK and p38 MAPK. CONCLUSIONS: RRF significantly inhibited MIRI through anti-oxidative, anti-inflammatory, and anti-apoptosis effects, and mechanisms were associated with its inhibition on phosphorylation of JNK and p38 MAPK.


Asunto(s)
Flavonoides/uso terapéutico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Rosa/química , Animales , Apoptosis/efectos de los fármacos , Flavonoides/farmacología , Inflamación/tratamiento farmacológico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/prevención & control , Fosforilación/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos
17.
Dev Comp Immunol ; 73: 21-26, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28263837

RESUMEN

Dendritic cells (DCs) comprise a system of highly professional antigen presenting cells (APCs) which connect innate and adaptive immunity by undergoing dramatic shift in their maturation state. Phytomedicine Echinacea purpurea extracts (EE) could modulate murine dendritic cell fate and function. However, the underlying mechanism of EE on DCs development and maturation remains limited. In this study, immature DCs were induced phenotypic maturation with up-regulated expression of key accessory molecules and the phagocytic activity was decreased after being treated with EE (400 µg/ml) for 48 h. We found that TLR1/2, JNK, p38-MAPK and NF-κB pathways were activated following EE exposure. Notably, JNK activation was demonstrated to be associated with increased IFN-γ response while p38-MAPK pathway exhibited immuno-regulatory effects via induction of IL-10 and TGF-ß1. Furthermore, it was verified that NF-κB signaling was responsible for EE-induced synthesis of IFN-γ, IL-12 and TGF-ß1, but not for IL-10 induction. These results indicate that EE have the immunomodulatory potency to promote both phenotypic and functional maturation of BMDCs via modulating the activation of JNK, p38-MAPK and NF-κB pathways. Our findings contributed to the current understanding of the immunoregulatory function of EE and the mechanism of DCs maturation.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Echinacea , Extractos Vegetales/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Células Dendríticas/citología , Células Dendríticas/inmunología , Femenino , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , FN-kappa B/efectos de los fármacos , Transducción de Señal/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos
18.
Med Sci Monit ; 23: 1448-1455, 2017 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-28341822

RESUMEN

BACKGROUND At present, the treatment of coxsackievirus-induced myocarditis remains difficult. Berberine (BBR), an isoquinoline alkaloid isolated from traditional medicine herbs, exhibits significant anti-viral efficacy against various viruses. However, the underlying mechanism by which BBR controls CVB3 infection has not yet been reported. The purpose of this study was to investigate the anti-viral efficacy of BBR against CVB3 infection and its mechanism. MATERIAL AND METHODS In our experiments, the protein levels of VP1 and MAPKs signal pathway were measured by Western blot. The mRNA level of VP1 was measured by RT-PCR. The virus titers were determined by TCID50 assay. RESULTS We found that BBR treatment significantly decreased CVB3 replication in HeLa cells. In addition, the BBR treatment reduced the phosphorylation levels of JNK and p38 MAPK upon CVB3 infection in both HeLa cells and primary rat myocardial cells. CONCLUSIONS Taken together, these results suggest that BBR inhibits CVB3 replication through the suppression of JNK and p38 MAPK activation, shedding new light on the investigation of therapeutic strategies against CVB3-induced viral myocarditis.


Asunto(s)
Berberina/metabolismo , Berberina/uso terapéutico , Enterovirus Humano B/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Antivirales/farmacología , Técnicas de Cultivo de Célula , Infecciones por Coxsackievirus , Células HeLa , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Miocarditis/tratamiento farmacológico , Miocitos Cardíacos/metabolismo , Fosforilación , Cultivo Primario de Células , Ratas , Receptores Virales/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Inflammation ; 40(3): 788-797, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28194607

RESUMEN

Exposure to ozone has been associated with airway inflammation and glucocorticoid insensitivity. This study aimed to observe the capacity of anti-murine interleukin-17A monoclonal antibody (IL-17mAb) to reverse ozone-induced glucocorticoid insensitivity and to detect its effects with glucocorticoids in protecting against airway inflammation. After C57/BL6 mice were exposed to ozone (2.5 ppm; 3 h) for 12 times over 6 weeks, PBS, IL-17mAb (50 ug/ml), dexamethasone (2 mg/kg), and combination administration of IL-17mAb (50 ug/ml) and dexamethasone (2 mg/kg) were intraperitoneally injected into mice at a dose of 0.1 ml, respectively, for 10 times over 5 weeks. At sacrifice, lung histology, airway inflammatory cells, levels of related cytokines in bronchoalveolar lavage fluid (BALF), and serum were analyzed, airway inflammatory cell infiltration density and mean linear intercept (Lm) were measured, the expression of IL-17A mRNA, glucocorticoid receptors (GR), NF-κB, and p38 mitogen-activated protein kinase (MAPK) phosphorylation were determined. We found that combination administration markedly reduced ozone-induced total inflammatory cells, especially neutrophils; inhibited levels of cytokines, including IL-8, IL-17A, and TNF-α in BALF; and suppressed airway inflammatory cell infiltration density and Lm. Additionally, combination administration significantly elevated levels of IFN-γ in BALF, decreased the dexamethasone-induced increase of IL-17A mRNA, and increased the expression of GR and decrement of NF-κB and p38MAPK phosphorylation, which are also related to glucocorticoids insensitivity. Collectively, combination administration shows profound efficacy in inhibiting certain cytokines, and IL-17 mAb partly improved the glucocorticoids insensitivity via modulating the enhanced production rate and improving expression of IL-17A induced by glucocorticoids administration and p38MAPK, NF-κB signaling pathway.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Glucocorticoides/farmacología , Interleucina-17/inmunología , Ozono/toxicidad , Animales , Anticuerpos Monoclonales/farmacología , Asma/tratamiento farmacológico , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Dexametasona/farmacología , Dexametasona/uso terapéutico , Resistencia a Medicamentos/efectos de los fármacos , Quimioterapia Combinada , Glucocorticoides/uso terapéutico , Ratones , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Neutrófilos/inmunología , Neutrófilos/patología , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
Anatol J Cardiol ; 17(5): 367-373, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28100897

RESUMEN

OBJECTIVE: Xindening oral liquid (Xin) is a widely used traditional Chinese medicine for the treatment of chronic heart failure (CHF). However, the exact mechanisms related to its therapeutic effects against CHF remain unclear. In the present study, we investigate the effects of Xin on cardiac function in CHF rats and the possible mechanisms involved. METHODS: Transverse aortic constriction (TAC) was conducted to induce a CHF rat model in this study. Sixty male Wistar rats were randomly assigned to six groups 28 days after TAC: sham; CHF model; Xin at concentrations of 5 ml/kg, 10 mL/kg, and 20 mL/kg; and QiLi 0.6 g/kg. After four weeks, the rats were treated with Xin (5, 10, or 20 mL/kg/d) for six weeks consecutively. At the end of the study, the cardiac function, heart weight index (HWI) and left ventricular mass index (LVMI), serum level of LDH, B-type natriuretic peptide (BNP), cTnI and CK-MB, and collagen volume fraction were studied. The expression of transforming growth factor-ß1 (TGF-ß1), drosophila mothers against decapentaplegic protein 3 (Smad3), and p38 mitogen activated protein kinase (p38 MAPK) were detected. RESULTS: The results showed that Xin treatment significantly improved cardiac function but decreased the serum level of LDH, BNP, cTnI, and CKMB of CHF rats. In addition, it reduced the HWI, LVMI, and collagen volume fraction compared with the model group. Xin treatment significantly improved cardiac function and attenuated cardiac fibrosis by suppressing the p38 MAPK and TGF-ß1/Smad3 signaling pathway in CHF rats. CONCLUSION: These results suggested that Xin might be a promising complementary treatment for CHF. More detailed experimental studies will be carried out in our subsequent research.


Asunto(s)
Antagonistas de Receptores Adrenérgicos beta 1/farmacología , Medicamentos Herbarios Chinos/farmacología , Insuficiencia Cardíaca/tratamiento farmacológico , Practolol/farmacología , Administración Oral , Antagonistas de Receptores Adrenérgicos beta 1/administración & dosificación , Antagonistas de Receptores Adrenérgicos beta 1/uso terapéutico , Animales , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/uso terapéutico , Hemodinámica/efectos de los fármacos , Masculino , Practolol/administración & dosificación , Practolol/uso terapéutico , Ratas , Ratas Wistar , Factor de Crecimiento Transformador beta1/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA