Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.785
Filtrar
1.
Chem Biol Interact ; 401: 111164, 2024 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-39111524

RESUMEN

Ganoderic Acid A (GAA) has demonstrated beneficial effects in anti-inflammatory and anti-oxidative stress studies. However, it remains unknown whether GAA exerts positive impacts on bone loss induced by lipopolysaccharide (LPS). This study aims to investigate the influence of GAA on bone loss in LPS-treated rats. The study assesses changes in the viability and osteogenic potential of MC3T3-E1 cells, as well as osteoclast differentiation in RAW264.7 cells in the presence of LPS using CCK-8, ALP staining, AR staining, and Tartrate-resistant acid phosphatase (TRAP) staining. In vitro experiments indicate that LPS-induced inhibition of osteoclasts (OC) and Superoxide Dismutase 2 (SOD2) correlates with heightened levels of inflammation and oxidative stress. Furthermore, GAA has displayed the ability to alleviate oxidative stress and inflammation, enhance osteogenic differentiation, and suppress osteoclast differentiation. Animal experiment also proves that GAA notably upregulates SOD2 expression and downregulates TNF-α expression, leading to the restoration of impaired bone metabolism, improved bone strength, and increased bone mineral density. The collective experimental findings strongly suggest that GAA can enhance osteogenic activity in the presence of LPS by reducing inflammation and oxidative stress, hindering osteoclast differentiation, and mitigating bone loss in LPS-treated rat models.


Asunto(s)
Diferenciación Celular , Ácidos Heptanoicos , Inflamación , Lanosterol , Lipopolisacáridos , Osteoclastos , Osteogénesis , Estrés Oxidativo , Ratas Sprague-Dawley , Superóxido Dismutasa , Animales , Lipopolisacáridos/farmacología , Estrés Oxidativo/efectos de los fármacos , Masculino , Ratones , Ratas , Células RAW 264.7 , Superóxido Dismutasa/metabolismo , Inflamación/metabolismo , Inflamación/tratamiento farmacológico , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Diferenciación Celular/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Lanosterol/análogos & derivados , Lanosterol/farmacología , Lanosterol/uso terapéutico , Ácidos Heptanoicos/farmacología , Ácidos Heptanoicos/uso terapéutico , Densidad Ósea/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Resorción Ósea/prevención & control , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/metabolismo
2.
Dokl Biochem Biophys ; 518(1): 361-371, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39023671

RESUMEN

Stroke is recognized as a leading cause of disability and mortality worldwide, posing a significant challenge, particularly in developing countries. The current study aimed to evaluate the neuroprotective effect of Ganoderic acid (GA) against focal ischemic stroke in rats. MATERIAL AND METHODS: Swiss Wistar rats were used for the current study. The rats were subjected to middle cerebral artery occlusion (MCAO) to simulate transient focal ischemia, followed by reperfusion. Various neurological parameters, including infarct size, neurological deficit score, brain water content, Evans blue leakage, nitric oxide (NO), inducible nitric oxide synthase (iNOS), lactate dehydrogenase (LDH), antioxidant levels, inflammatory cytokines, apoptosis markers, inflammatory parameters, and matrix metalloproteinases (MMP) levels, were estimated. Additionally, mRNA expressions were evaluated in the brain tissue. RESULTS: Dose dependently treatment of GA significantly (P < 0.001) suppressed the infarct size, neurological deflects score, brain water, evans blue leakage, NO, iNOS, LDH, C-X-C chemokine receptor type 4 (CXCR-4), monocyte chemoattractant protein-1 (MCP-1), S100 calcium-binding protein B (S-100ß) and K+-Cl- cotransporter 1 (KCC1) positive cells. GA altered the level of oxidative stress parameters like Total antioxidant capacity (T-AOC), 8-hydroxy-2'-deoxyguanosine (8-OhdG), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione (GSH), malonaldehyde (MDA); cytokines viz., tumor necrosis factor-α (TNF-α), interleukin-1 (IL-1), IL-1ß, IL-6, IL-9, IL-10; inflammatory parameters such as inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), prostaglandin (PGE2), Nuclear factor kappa B (NF-κB); apoptosis parameters like B-cell leukemia/lymphoma 2 protein (Bcl-2), Bcl-2-associated protein x (Bax), Caspase-3; matrix metallopeptidase (MMP) parameters like MMP-2, MMP-3, and MMP-9, respectively. GA remarkably suppressed the mRNA expression of TRL-4, Syndecan-1, CSF, Aquaporin-1, OCT3, and RFX1. CONCLUSION: Ganoderic acid exhibited the protection against the cerebral ischemia reperfusion via multiple mechanism.


Asunto(s)
Infarto de la Arteria Cerebral Media , Fármacos Neuroprotectores , Estrés Oxidativo , Ratas Wistar , Animales , Estrés Oxidativo/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Ratas , Masculino , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/metabolismo , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Accidente Cerebrovascular Isquémico/patología , Accidente Cerebrovascular Isquémico/metabolismo , Lanosterol/análogos & derivados , Lanosterol/farmacología , Lanosterol/uso terapéutico , Ácidos Heptanoicos/farmacología , Ácidos Heptanoicos/uso terapéutico , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Antioxidantes/farmacología
3.
Clin Med (Lond) ; 24(3): 100217, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38710328

RESUMEN

Statin-induced immune-mediated necrotising myopathy (IMNM) is an inflammatory myopathy that can present as proximal muscle weakness and, in some cases, as dysphagia and respiratory distress. In this report, we present a case of statin-induced IMNM in a 78-year-old male. The patient had significantly high levels of creatinine kinase and myoglobinuria and experienced gradual weakness in the proximal muscles for 1 month after initiating a 20 mg dose of Atorvastatin 10 months before admission. Rapid clinical improvement was observed with the use of high-dose glucocorticoids in conjunction with methotrexate.


Asunto(s)
Atorvastatina , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Miositis , Humanos , Masculino , Anciano , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Miositis/inducido químicamente , Miositis/tratamiento farmacológico , Atorvastatina/uso terapéutico , Atorvastatina/efectos adversos , Metotrexato/uso terapéutico , Metotrexato/efectos adversos , Pirroles/efectos adversos , Pirroles/uso terapéutico , Inmunosupresores/uso terapéutico , Inmunosupresores/efectos adversos , Ácidos Heptanoicos/efectos adversos , Ácidos Heptanoicos/uso terapéutico , Necrosis/inducido químicamente , Glucocorticoides/uso terapéutico , Enfermedades Musculares/inducido químicamente , Enfermedades Musculares/tratamiento farmacológico
5.
J Pharm Pharmacol ; 76(4): 354-367, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38330446

RESUMEN

OBJECTIVES: Reportedly, ganoderic acid A (GA-A) increases the sensitivity of hepatocellular carcinoma cells to cisplatin (DDP) chemotherapy. Therefore, this study aims to fathom the influence of GA-A on lung cancer cells. METHODS: After the construction of A549/DDP cells through exposure to DDP, the effects of GA-A on A549 and A549/DDP cells were revealed by cellular functional assays, western blot and quantitative reverse transcription PCR (qRT-PCR). The DDP-resistant lung cancer tumor was established in vivo, followed by further validation of the mechanism of GA-A. RESULTS: GA-A suppressed the viability, migration, and invasion while downregulating Beclin and autophagy marker LC3II/LC3I levels and upregulating P62 levels in A549 and A549/DDP cells. These effects were reversed by circFLNA overexpression. Also, GA-A reinforced the sensitivity of A549/DDP cells to DDP, elevated the apoptosis and regulated the circFLNA/miR-486-3p/cytochrome P450 family 1 subfamily A member 1 (CYP1A1)/X-ray repair cross-complementing 1 (XRCC1) axis. The reversal effects of circFLNA overexpression on GA-A-induced viability and apoptosis of A549/DDP cells could all be counteracted in the presence of 3MA. GA-A inhibited lung cancer tumor growth and blocked autophagy. CONCLUSION: GA-A suppresses autophagy by regulating the circFLNA/miR-486-3p/CYP1A1/XRCC1 axis to strengthen the sensitivity of lung cancer cells to DDP.


Asunto(s)
Antineoplásicos , Autofagia , Carcinoma de Pulmón de Células no Pequeñas , Ácidos Heptanoicos , Lanosterol , Neoplasias Pulmonares , MicroARNs , Humanos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Autofagia/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular , Cisplatino/farmacología , Citocromo P-450 CYP1A1/efectos de los fármacos , Citocromo P-450 CYP1A1/metabolismo , Resistencia a Antineoplásicos , Ácidos Heptanoicos/farmacología , Ácidos Heptanoicos/uso terapéutico , Lanosterol/análogos & derivados , Lanosterol/farmacología , Lanosterol/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , MicroARNs/efectos de los fármacos , MicroARNs/metabolismo , ARN Circular/efectos de los fármacos , ARN Circular/metabolismo , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X/efectos de los fármacos , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X/metabolismo
6.
Eur Heart J Cardiovasc Pharmacother ; 10(4): 307-315, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-38196131

RESUMEN

AIMS: Objective methods to determine statin adherence are requested to improve lipid management. We have recently established a method to detect reduced adherence to atorvastatin therapy with cut-off values based on the sum of atorvastatin and its major metabolites in the blood. We aimed to validate this method in patients with and without cardiovascular disease, and optimize previous cut-off values. METHODS AND RESULTS: The pharmacokinetic study included 60 participants treated with atorvastatin 20 mg (N = 20), 40 mg (N = 20), and 80 mg (N = 20). Atorvastatin was then stopped and blood samples collected from day zero to day four. Quantification of the parent drug and its metabolites in blood plasma was performed with a liquid chromatography-tandem mass spectrometry assay. The cut-off values for reduced adherence were validated and optimized by calculating diagnostic sensitivity and specificity. Our candidate cut-off value of dose-normalized six-component sum of atorvastatin plus metabolites <0.10 nM/mg provided a sensitivity of 97% and a specificity of 93% for detecting ≥2 omitted doses. An optimized cut-off <0.062 nM/mg provided a sensitivity of 90% and a specificity of 100%. An alternative simplified two-component metabolite sum with a cut-off value <0.05 nM/mg provided a sensitivity of 98% and a specificity of 76%. An optimized cut-off <0.02 nM/mg provided a sensitivity of 97% and a specificity of 98%. CONCLUSION: This validation study confirms that our direct method discriminates reduced adherence from adherence to atorvastatin therapy with high diagnostic accuracy. The method may improve lipid management in clinical practice and serve as a useful tool in future studies.


Asunto(s)
Atorvastatina , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Cumplimiento de la Medicación , Atorvastatina/farmacocinética , Atorvastatina/uso terapéutico , Atorvastatina/sangre , Humanos , Masculino , Femenino , Persona de Mediana Edad , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacocinética , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/sangre , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Anciano , Ácidos Heptanoicos/farmacocinética , Ácidos Heptanoicos/administración & dosificación , Ácidos Heptanoicos/sangre , Ácidos Heptanoicos/uso terapéutico , Pirroles/farmacocinética , Pirroles/sangre , Pirroles/administración & dosificación , Espectrometría de Masas en Tándem , Cromatografía Liquida , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/prevención & control , Reproducibilidad de los Resultados , Relación Dosis-Respuesta a Droga
7.
Aging (Albany NY) ; 15(23): 13669-13679, 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-38048213

RESUMEN

Mild cognitive impairment (MCI) is a common symptom observed in people over 60 years old and is found to be aggravated by hypercholesterolemia. Severe neuroinflammation induced by BBB dysfunction and monocyte infiltration might be responsible for neuron damage and cognitive impairment. Atorvastatin is a lipid-lowering drug that is widely applied for the treatment of cardiovascular diseases. However, the potential function of Atorvastatin in hypercholesterolemia-induced MCI remains uncertain. Our research will explore the potential therapeutic function of Atorvastatin in memory deficits induced by chronic hypercholesterolemia. ApoE-/- mice were utilized to mimic the state of chronic hypercholesterolemia and were divided into four groups. Animals in the WT and ApoE-/-groups were orally administered with normal saline, while WT mice in the Atorvastatin group and ApoE-/- mice in the ApoE-/-+ Atorvastatin group were orally administered with 10 mg/kg/day Atorvastatin. Markedly increased plasma cholesterol levels reduced RI in the long-term memory test and the spatial short-term memory test, declined mobility in the open field test, and downregulated PSD-95 and BDNF were observed in ApoE-/- mice, all of which were signally reversed by Atorvastatin. Moreover, the percentages of brain Ly6Chi CD45+ cells and CD3+ CD45+ cells, as well as the blood Ly6Chi CD45+ cells, plasma IL-12/IL-23 levels and IL-17 level were found notably increased in ApoE-/- mice, all of which were largely repressed by Atorvastatin. Lastly, the increased BBB permeability, decreased ZO-1 and occludin levels, and reduced KLF2 level were markedly abolished by Atorvastatin. Collectively, Atorvastatin mitigated memory deficits and brain monocyte infiltration in ApoE-/- mice.


Asunto(s)
Ácidos Heptanoicos , Hipercolesterolemia , Hiperlipidemias , Humanos , Ratones , Animales , Atorvastatina/farmacología , Atorvastatina/uso terapéutico , Hipercolesterolemia/complicaciones , Hipercolesterolemia/tratamiento farmacológico , Monocitos/metabolismo , Ácidos Heptanoicos/farmacología , Ácidos Heptanoicos/uso terapéutico , Pirroles/farmacología , Hiperlipidemias/tratamiento farmacológico , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Encéfalo/metabolismo
8.
Adv Med Sci ; 68(2): 258-264, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37478516

RESUMEN

PURPOSE: For many years, statins have been the most commonly used drugs in cholesterol-lowering therapy. In addition to these therapeutic effects, statins exhibit other, pleiotropic effects that can be beneficial, but also harmful to cells and tissues. The aim of this research was to determine and compare the pleiotropic effects of structurally different statins: atorvastatin, simvastatin and rosuvastatin at different concentrations on hepatocellular carcinoma (HepG2) cells. MATERIALS AND METHODS: The MTT assay was used to determine the cytotoxic effects of statins. The influence of statins on the production of reactive oxygen species (ROS) was determined by measuring fluorescent response of 2,7-dichlorofluorescein diacetate (DCFH-DA). The effect of statins on glucose production and excretion was determined with glucose production assay. RESULTS: The obtained results confirmed that all tested statins exhibit cytotoxic effects, increase the production of ROS as well as the production and excretion of glucose from HepG2 cells. It was observed that all the mentioned effects are more pronounced with lipophilic statins, atorvastatin and simvastatin compared to hydrophilic rosuvastatin. CONCLUSION: The less pronounced pleiotropic effects of rosuvastatin on HepG2 cells are probably due to differences in structure and solubility compared to atorvastatin and simvastatin. Transporter-dependent and a slower influx of rosuvastatin into cells compared to the tested lipophilic statins probably lead to a weaker accumulation of rosuvastatin in HepG2 cells, which results in less pronounced pleiotropic effects compared to lipophilic atorvastatin and simvastatin.


Asunto(s)
Carcinoma Hepatocelular , Ácidos Heptanoicos , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Neoplasias Hepáticas , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Rosuvastatina Cálcica/farmacología , Atorvastatina/farmacología , Simvastatina/farmacología , Simvastatina/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Especies Reactivas de Oxígeno , Ácidos Heptanoicos/uso terapéutico , Pirroles/uso terapéutico , Fluorobencenos/uso terapéutico , Pirimidinas/uso terapéutico , Sulfonamidas/uso terapéutico , Neoplasias Hepáticas/tratamiento farmacológico , Glucosa
9.
Pharmacol Res Perspect ; 11(3): e01089, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37186070

RESUMEN

In this exploratory study from a randomized double-blinded crossover trial including 70 patients with coronary heart disease and self-perceived muscular side effects of statins, we aimed to determine the relationship between low-density lipoprotein cholesterol (LDL-C) reduction and atorvastatin metabolite plasma concentrations. All patients underwent a 7 weeks treatment period with atorvastatin 40 mg/day and a 7 weeks placebo period in random order. Nonlinear regression with a three-parameter equation explored the relationship between percentage LDL-C reduction (statin vs. placebo) and the pharmacokinetic variables. Mean LDL-C reduction was 49% (range 12% to 71%). The sum of 4-OH-atorvastatin acid and lactone correlated moderately with the LDL-C response (Spearman ρ 0.27, 95% confidence interval [CI]: 0.03 to 0.48). Accordingly, nonlinear regression showed R2 of 0.14 (95% CI: 0.03 to 0.37, R2 adjusted equaled 0.11). Even a perfect underlying correlation of 1.0 showed R2 = 0.32 by simulation, using historical intra-individual LDL-C variation (8.5%). The 90% inhibitory concentration was 2.1 nmol/L, and the 4-OH-metabolite sum exceeded this threshold in 34% of the patients. In conclusion, trough plasma concentrations of 4-OH-atorvastatin metabolites correlated moderately to the LDL-C reduction. A plateau LDL-C response was observed above a pharmacokinetic threshold, below which the response was highly variable. The usefulness of monitoring concentrations of atorvastatin metabolites to optimize the individual dosage have limitations, but its supportive potential may be pursued in relevant patient subsets to achieve adequate efficacy at the lowest possible dose. The results add knowledge to the overall understanding of the variable LDL-C response mediated by atorvastatin.


Asunto(s)
Anticolesterolemiantes , Enfermedad Coronaria , Ácidos Heptanoicos , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Humanos , Atorvastatina/uso terapéutico , LDL-Colesterol , Anticolesterolemiantes/uso terapéutico , Anticolesterolemiantes/farmacología , Ácidos Heptanoicos/farmacología , Ácidos Heptanoicos/uso terapéutico , Pirroles , Triglicéridos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Enfermedad Coronaria/tratamiento farmacológico , Enfermedad Coronaria/inducido químicamente
10.
Clin Sci (Lond) ; 135(16): 1945-1980, 2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34401905

RESUMEN

Eicosanoids comprise a group of oxidation products of arachidonic and 5,8,11,14,17-eicosapentaenoic acids formed by oxygenases and downstream enzymes. The two major pathways for eicosanoid formation are initiated by the actions of 5-lipoxygenase (5-LO), leading to leukotrienes (LTs) and 5-oxo-6,8,11,14-eicosatetraenoic acid (5-oxo-ETE), and cyclooxygenase (COX), leading to prostaglandins (PGs) and thromboxane (TX). A third group (specialized pro-resolving mediators; SPMs), including lipoxin A4 (LXA4) and resolvins (Rvs), are formed by the combined actions of different oxygenases. The actions of the above eicosanoids are mediated by approximately 20 G protein-coupled receptors, resulting in a variety of both detrimental and beneficial effects on airway smooth muscle and inflammatory cells that are strongly implicated in asthma pathophysiology. Drugs targeting proinflammatory eicosanoid receptors, including CysLT1, the receptor for LTD4 (montelukast) and TP, the receptor for TXA2 (seratrodast) are currently in use, whereas antagonists of a number of other receptors, including DP2 (PGD2), BLT1 (LTB4), and OXE (5-oxo-ETE) are under investigation. Agonists targeting anti-inflammatory/pro-resolving eicosanoid receptors such as EP2/4 (PGE2), IP (PGI2), ALX/FPR2 (LXA4), and Chemerin1 (RvE1/2) are also being examined. This review summarizes the contributions of eicosanoid receptors to the pathophysiology of asthma and the potential therapeutic benefits of drugs that target these receptors. Because of the multifactorial nature of asthma and the diverse pathways affected by eicosanoid receptors, it will be important to identify subgroups of asthmatics that are likely to respond to any given therapy.


Asunto(s)
Antiasmáticos/uso terapéutico , Antiinflamatorios/uso terapéutico , Asma/tratamiento farmacológico , Receptores Eicosanoides/agonistas , Receptores Eicosanoides/antagonistas & inhibidores , Acetatos/farmacología , Acetatos/uso terapéutico , Animales , Antiasmáticos/farmacología , Antiinflamatorios/farmacología , Asma/metabolismo , Asma/fisiopatología , Benzoquinonas/farmacología , Benzoquinonas/uso terapéutico , Biomarcadores/metabolismo , Ciclopropanos/farmacología , Ciclopropanos/uso terapéutico , Ácidos Heptanoicos/farmacología , Ácidos Heptanoicos/uso terapéutico , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/fisiopatología , Ratones , Quinolinas/farmacología , Quinolinas/uso terapéutico , Receptores Eicosanoides/efectos de los fármacos , Sulfuros/farmacología , Sulfuros/uso terapéutico
11.
Inflammation ; 44(5): 1908-1915, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34037898

RESUMEN

The aim of this study is to investigate the effects of ganoderic acid A (GAA) on OVA-induced asthma in mice. Mouse asthma model was established by ovalbumin (OVA) in vitro. Diff-Quik staining was used to observe the total numbers of cells and the number of classification cells in each group, and HE staining was used to observe lung inflammation in lung tissue sections. ELISA was used to detect the effect of GAA on the levels of interleukin-4 (IL-4), IL-5, and IL-13 in serum and lung tissue. The expression levels of TLR/NF-κB were detected by Western blot. Immunohistochemistry was used to observe the expression changes of TLR4 and P-P65. Compared with the normal group, the inflammatory cell count, IL-4, IL-5, and IL-13 expression in the model group increased, and TLR/NF-kB signal protein expression increased. Compared with the model group, in GAA group, the number of inflammatory cells, the expression of IL-4, IL-5, and IL-13 decreased, and the expression of TLR/NF-kB signaling protein decreased. GAA regulated lung inflammation in asthmatic mice by inhibiting TLR/NF-kB signaling pathway.


Asunto(s)
Asma/inducido químicamente , Asma/tratamiento farmacológico , Ácidos Heptanoicos/uso terapéutico , Mediadores de Inflamación/antagonistas & inhibidores , Lanosterol/análogos & derivados , Ovalbúmina/toxicidad , Animales , Asma/metabolismo , Citocinas/antagonistas & inhibidores , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Ácidos Heptanoicos/farmacología , Mediadores de Inflamación/metabolismo , Lanosterol/farmacología , Lanosterol/uso terapéutico , Ratones , Ratones Endogámicos BALB C , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/metabolismo
12.
Chem Biol Interact ; 344: 109529, 2021 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-34029542

RESUMEN

Ganoderic acid A (GAA), one of the major triterpenoid components extracted from Ganoderma mushroom has been shown to possess numerous important pharmacological activities. The present study was aimed to investigate the mechanisms of GAA on carbon tetrachloride (CCl4)-induced kidney inflammation, fibrosis and oxidative stress in mice. The male mice were treated with 25 and 50 mg/mg GAA after stimulated with CCl4. Our results showed that GAA improved renal damage by decreasing the serum levels of creatinine, urea, uric acid and alleviating kidney fibrosis. GAA ameliorated CCl4-induced indices of inflammation. GAA suppressed oxidative stress by regulating the glutathione antioxidant system and the thioredoxin antioxidant system. GAA increased the activations of thioredoxin reductase (TrxR), Trx, GSH, SOD, GPx. Furthermore, GAA supplementation inhibited the JAK and STAT3 pathway. GAA inhibited the activations of RhoA, ROCK, NF-κB, TGF-ß and Smad3. Thus, this study demonstrated that GAA possesses immune-protective properties through regulating the Trx/TrxR, JAK2/STAT3 and RhoA/ROCK pathways.


Asunto(s)
Antiinflamatorios/uso terapéutico , Antioxidantes/uso terapéutico , Fibrosis/tratamiento farmacológico , Ácidos Heptanoicos/uso terapéutico , Enfermedades Renales/tratamiento farmacológico , Lanosterol/análogos & derivados , Transducción de Señal/efectos de los fármacos , Animales , Tetracloruro de Carbono , Fibrosis/inducido químicamente , Fibrosis/patología , Janus Quinasa 2/metabolismo , Enfermedades Renales/inducido químicamente , Enfermedades Renales/patología , Lanosterol/uso terapéutico , Masculino , Ratones Endogámicos ICR , Estrés Oxidativo/efectos de los fármacos , Proteína smad3/metabolismo , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Tiorredoxinas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Quinasas Asociadas a rho/metabolismo
13.
Biochem Pharmacol ; 188: 114561, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33857491

RESUMEN

Major depressive disorder (MDD) is a common, chronic, recurrent disease. The existing drugs are ineffective for approximately half of patients, so the development of antidepressant drugs with novel mechanisms is urgent. Cumulative evidence has shown neuro-inflammation plays a key role in the etiology of major depressive disorder. Clinical studies implicated that bile acids, an important component of gut-brain axis, inhibit neuro-inflammation and mediate the pathophysiology of the MDD. Here, we found that ganoderic acid A (GAA) modulated bile acid receptor FXR (farnesoid X receptor), inhibited brain inflammatory activity, and showed antidepressant effects in the chronic social defeat stress depression model, tail suspension, forced swimming, and sucrose preference tests. GAA directly inhibited the activity of the NLRP3 inflammasome, and activated the phosphorylation and expression of the AMPA receptor by modulating FXR in the prefrontal cortex of mice. If we knocked out FXR or injected the FXR-specific inhibitor z-gugglesterone (GS), the antidepressant effects induced by GAA were completely abolished. These results suggest that GAA modulates the bile acid receptor FXR and subsequently regulates neuroimmune and antidepressant behaviors. GAA and its receptor FXR have potential as targets for the treatment of MDD.


Asunto(s)
Antidepresivos/uso terapéutico , Depresión/metabolismo , Ácidos Heptanoicos/uso terapéutico , Lanosterol/análogos & derivados , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Receptores Citoplasmáticos y Nucleares/deficiencia , Sinapsis/metabolismo , Animales , Antidepresivos/farmacología , Depresión/tratamiento farmacológico , Depresión/psicología , Ácidos Heptanoicos/farmacología , Lanosterol/farmacología , Lanosterol/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Receptores Citoplasmáticos y Nucleares/genética , Derrota Social , Sinapsis/efectos de los fármacos , Sinapsis/genética
14.
Biochem Pharmacol ; 185: 114422, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33482151

RESUMEN

Multiple sclerosis (MS), as an inflammatory demyelinating disorder of central nervous system, is the leading cause of non-traumatic neurologic disability in young adults. The pathogenesis of MS remains unknown, however, a dysregulation of glia-neuroimmune signaling plays a key role during progressive disease stage. Most of the existing drugs are aimed at the immune system, but there is no approved drug by promoting remyelination after demyelination so far. There is a great interest in identifying novel agents for treating MS bytargeting to switch the immune imbalance from pro-inflammation and apoptosis to anti-inflammation and regeneration during remyelination phase. Here, we reported that ganoderic acid A (GAA) significantly enhanced the remyelination and rescued motor deficiency in two animal models of MS, including cuprizone-induced demyelination and myelin oligodendrocyte glycoprotein (MOG) 35-55-induced experimental autoimmune encephalomyelitis model. In these two independent MS animal models, GAA modulated neuroimmune to enhance the anti-inflammatory and regeneration markers IL-4 and BDNF, inhibited inflammatory markers IL-1ß and IL-6, followed by down-regulation of microglia activation and astrocyte proliferation. Pharmacological and genetic ablation of farnesoid-X-receptor (FXR) abolished GAA-induced remyelination and restoration of motor deficiency in MS mice. Thus, GAA is a novel and potential therapeutic agent that can rescue MS neuroimmune imbalance and remyelination through an FXR receptor-dependent mechanism. Clinical investigation on the therapeutic effect of GAA in improving remyelination of the MS patients to rescue the motor function is warranted.


Asunto(s)
Antiinflamatorios/uso terapéutico , Ácidos Heptanoicos/uso terapéutico , Lanosterol/análogos & derivados , Esclerosis Múltiple/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Regeneración/fisiología , Remielinización/fisiología , Animales , Antiinflamatorios/farmacología , Femenino , Ácidos Heptanoicos/farmacología , Lanosterol/farmacología , Lanosterol/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/patología , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Regeneración/efectos de los fármacos , Remielinización/efectos de los fármacos
15.
Immunol Lett ; 226: 1-6, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32565114

RESUMEN

The purpose of the experiment was to explore the effect of Ganoderic acid A (GAA) on adjuvant-induced arthritis in rats. In this study, the rat model of collagen-induced rheumatoid arthritis (CIA) was established with type II collagen plus Freund's complete adjuvant. Arthritis index, joint pathology, toe swelling, hemorheology, synovial cell apoptosis, related cytokines and JAK3/STAT3 and nuclear factor-κB (NF-κB) signaling pathway were measured in rats. We found that GAA can significantly inhibit the arthritis index, improve joint pathology, reduce toe swelling, improve blood rheology, improve synovial cell apoptosis, and restore related cytokine negative regulation JAK3/STAT3 and NF-κB signaling pathways. In conclusion, GAA has an obvious therapeutic effect on joint inflammation of toes in CIA model rats, which may be due to the regulation of JAK3/STAT3 and NF-κB signaling pathway.


Asunto(s)
Antiinflamatorios/uso terapéutico , Artritis Experimental/tratamiento farmacológico , Artritis Reumatoide/tratamiento farmacológico , Ácidos Heptanoicos/uso terapéutico , Lanosterol/análogos & derivados , Sinoviocitos/patología , Animales , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Janus Quinasa 3/metabolismo , Lanosterol/uso terapéutico , Masculino , FN-kappa B/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
16.
Int Immunopharmacol ; 85: 106625, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32485356

RESUMEN

Triple-negative breast cancer (TNBC) has a more aggressive phenotype and higher metastasis and recurrence rates than other breast cancer subtypes. The immune microenvironment and hypoxic microenvironment of breast cancer constitute the survival environment of cancer cells, which is an important environment to support cancer cells. LXA4 and its analog, BML-111 is an important regulator of inflammatory cytokines, which provides a possible way for the treatment of inflammatory-related tumors. Here, in the in vitro experiment, we showed that BML-111 could inhibit the EMT and migration of TAMs-stimulated TNBC by down-regulating ILK as well as p-Akt and p-GSK3ß. And it could prevent the formation of breast cancer cell clusters. In the in vivo experiment, BML-111 could inhibit the metastasis of 4T1 breast cancer cells. We also demonstrated that BML-111 could affect macrophages in tumor microenvironment to prevent metastasis. These results showed that BML-111 could be a possible candidate for breast cancer therapy by targeting ILK and TAMs.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Ácidos Heptanoicos/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Ácidos Heptanoicos/uso terapéutico , Humanos , Interleucina-8/metabolismo , Pulmón/patología , Macrófagos/efectos de los fármacos , Ratones Endogámicos BALB C , Metástasis de la Neoplasia/prevención & control , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Microambiente Tumoral , Macrófagos Asociados a Tumores , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Int Immunopharmacol ; 84: 106543, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32353688

RESUMEN

This study aimed to investigate the protective effect of GanodericacidA (GA) on myocardial ischemia-reperfusion (MIR) injury. The myocardial injury model in rats was established by ligating left anterior descending coronary artery. We measured cardiac hemodynamic, antioxidant enzyme activity, and various biochemical indexes of myocardial tissue, and evaluated myocardial infarction and damage. Further, the expression of JAK2/STAT3/NF-κB signaling pathway-related proteins in myocardial tissue was measured by western blot. The results showed that the myocardial infarction extention was obviously reduced upon GA treatment. Compared with the control group, ischemia-reperfusion rats showed significant increase in lactate dehydrogenase (LDH) and creatine Kinase (CK), which were significantly decreased in GA group. Besides, GA pretreatment effectively decreased the levels of inflammatory cytokines in serum. The phosphorylation of Janus Kinase 2 (JAK2), signal transducer and activator of transcription (STAT3)and Nuclear factor-κB (NF-κB) in reperfusion group were significantly higher than that in control group, which were reversed upon GA treatment. In conclusion, GA may reduce myocardial injury by regulating JAK2/STAT3/NF-κB pathway.


Asunto(s)
Ácidos Heptanoicos/uso terapéutico , Janus Quinasa 2/metabolismo , Lanosterol/análogos & derivados , Daño por Reperfusión Miocárdica/tratamiento farmacológico , FN-kappa B/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Citocinas/sangre , Ácidos Heptanoicos/farmacología , Lanosterol/farmacología , Lanosterol/uso terapéutico , Masculino , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Función Ventricular Izquierda/efectos de los fármacos
18.
Pharmacology ; 105(9-10): 568-575, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31940650

RESUMEN

BACKGROUND: To study the protective effects of ganoderic acid A (GAA) on bleomycin (BLM)-induced pulmonary fibrosis. METHODS: ICR mice were intratracheally instilled with BLM to induce pulmonary fibrosis on day 0. Then the mice were orally given GAA (25, 50 mg/kg) or dexamethasone (2 mg/kg). After treatment for 21 days, the mice were sacrificed. Wet dry weight (W/D) ratio of lung was used to detect pulmonary edema. Myeloperoxidase (MPO), interleukin-1ß (IL-1ß), IL-6, tumor necrosis factor-α (TNF-α), malondialdehyde (MDA), and superoxide dismutase (SOD) were detected by enzyme-linked immunosorbent assay. Hematoxylin and eosin staining was used to evaluate the pathological changes. The levels of transforming growth factor ß (TGF-ß), phosphorylated-smad3 (p-smad3), p-IκB, and p-nuclear factor-kappa B (NF-κB) in lung tissue were detected by western blot. RESULTS: GAA treatment significantly improved MPO activity, W/D ratio, and lung histopathology. The protective effect of GAA may be related to downregulation of TNF-α, IL-1ß, IL-6, MDA and upregulation of SOD. In addition, GAA significantly decreased the levels of TGF-ß, p-smad3, p-IκB, and p-NF-κB, compared with those in BLM group. CONCLUSION: GAA has protective effect on BLM-induced lung injury, and TGF-ß/Smad-3/NF-κB signaling pathway may play an important role in the pathogenesis of BLM-induced lung injury.


Asunto(s)
Ácidos Heptanoicos/farmacología , Lanosterol/análogos & derivados , Pulmón/efectos de los fármacos , Extractos Vegetales/farmacología , Sustancias Protectoras/farmacología , Fibrosis Pulmonar/tratamiento farmacológico , Animales , Bleomicina/toxicidad , Citocinas/sangre , Ácidos Heptanoicos/uso terapéutico , Lanosterol/farmacología , Lanosterol/uso terapéutico , Pulmón/metabolismo , Pulmón/patología , Masculino , Malondialdehído/sangre , Ratones , Ratones Endogámicos ICR , FN-kappa B/metabolismo , Peroxidasa/metabolismo , Fitoterapia , Extractos Vegetales/administración & dosificación , Extractos Vegetales/uso terapéutico , Sustancias Protectoras/uso terapéutico , Edema Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/inducido químicamente , Proteína smad3/metabolismo , Superóxido Dismutasa/sangre , Factor de Crecimiento Transformador beta/metabolismo
19.
J Diabetes Complications ; 34(3): 107480, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31902653

RESUMEN

AIMS: To examine whether addition of amlodipine (5 mg)/atorvastatin (10 mg) A/A to Therapeutic Lifestyle change intervention (TLC) would beneficially modulate Metabolic Syndrome (MetS) and oxidized low-density lipoprotein (Ox-LDL) levels. METHODS: Patients with MetS (n = 53) were randomized to TLC + placebo or TLC + A/A for 12 months. Anthropometric measurements, blood pressure (BP), lipid profile, plasma Ox-LDL, and area under the curve of free fatty acid (AUCFFA) during oral glucose tolerance test, a marker of adipose tissue health, were assessed before and after the intervention. RESULTS: Twenty-six patients completed the study with an overall improvement of MetS (p = 0.02). TLC + placebo was beneficial in reversing MetS comparable to TLC + A/A (54% vs. 39%; p = 0.08). Both treatments decreased systolic BP (p ≤ 0.01). TLC + A/A also decreased diastolic BP and triglyceride levels. The changes in Ox-LDL levels directly correlated with changes in weight in the TLC-placebo group (r = 0.64; p = 0.04). AUCFFA determined the loss of fat mass (r = 0.472, p = 0.03). CONCLUSIONS: 1) Addition of A/A has the advantage of improving the lipid profile and BP; but TLC alone was comparable to TLC + A/A in improving MetS; 2) weight change determines the TLC-associated change in Ox-LDL levels; and 3) AT metabolic health is a significant predictor of TLC-associated loss of body fat mass.


Asunto(s)
Amlodipino/uso terapéutico , Terapia Conductista/métodos , Ácidos Heptanoicos/uso terapéutico , Síndrome Metabólico/terapia , Pirroles/uso terapéutico , Adulto , Anciano , Amlodipino/administración & dosificación , Atorvastatina/administración & dosificación , Biomarcadores/sangre , Presión Sanguínea/fisiología , Factores de Riesgo Cardiometabólico , Terapia Combinada , Combinación de Medicamentos , Femenino , Humanos , Estilo de Vida , Lípidos/sangre , Lipoproteínas LDL/sangre , Masculino , Síndrome Metabólico/sangre , Síndrome Metabólico/fisiopatología , Persona de Mediana Edad , Estrés Oxidativo/fisiología , Placebos , Conducta de Reducción del Riesgo
20.
Int Immunopharmacol ; 79: 106108, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31881376

RESUMEN

Diabetic nephropathy (DN) is the most paradigmatic complication of diabetes mellitus (DM) and brings about severe social and economic burdens. BML-111 is a potent agonist of Lipoxin A4 and has shown anti-inflammatory function in many diseases. The aim of the study is to investigate the effects of BML-111 on high glucose (HG) -induced mesangial cells. HBZY-1 cells were stimulated by HG with or without BML-111. ML385 was used as an Nrf2 inhibitor. Cell proliferation was measured by CC-K 8 assay. Besides, levels of TNF-α, IL-1, IL-6 and MCP-1 were detected by corresponding ELISA kits. DCFH-DA staining and an available ROS kit were employed to determine the ROS generation. In addition, extracellular matrix (ECM) accumulation was evaluated by immunofluorescence assay and western blot analysis. The protein expressions involved in Nrf2/HO-1 and MAPK pathway were assessed by western blot assay. Results indicated that BML-111 extremely inhibited HBZY-1 cell proliferation induced by HG. Moreover, BML-111 reduced the levels of TNF-α, IL-1, IL-6 and MCP-1, declined intracellular ROS level, and attenuated expression of ECM proteins laminin, fibronectin, collagen IV and TGF-ß1. In addition, BML-111 promoted the activation of Nrf2, HO-1, and NQO1, while suppressed the phosphorylation of p38 and JNK. Further, NRF2 silence reversed the inhibitory effects of BML-111 on HG-induce inflammation, oxidative stress and ECM accumulation, accelerate the MAPK signaling, and diminished the expression of Nrf2 pathway. In summary, BML-111 alleviated HG-induced injury in HBZY-1 cells by repressing inflammatory response, oxidative stress and ECM accumulation via activating Nrf2 and inhibiting MAPK pathway.


Asunto(s)
Antiinflamatorios/uso terapéutico , Nefropatías Diabéticas/tratamiento farmacológico , Ácidos Heptanoicos/uso terapéutico , Inflamación/tratamiento farmacológico , Células Mesangiales/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Línea Celular , Matriz Extracelular/efectos de los fármacos , Glucosa/metabolismo , Humanos , Células Mesangiales/patología , Terapia Molecular Dirigida , Estrés Oxidativo/efectos de los fármacos , Ratas , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA