Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.054
Filtrar
1.
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
2.
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
3.
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
4.
Anticancer Agents Med Chem ; 23(7): 832-838, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36330648

RESUMEN

BACKGROUND: Various natural products have been demonstrated for their anti-tumor activities. As a natural triterpenoid, the effects of ganoderic acid A on oxaliplatin chemotherapy for cancer treatment remain unclear. METHODS: A xenograft mouse model of colon cancer was constructed using the HT-29 cells. Ganoderic acid A was intravenously administered with or without oxaliplatin. The CCK-8 method was performed to assess cell viability. Flow cytometry was used to determine cell apoptosis and subtyping of T cells. Cytotoxicity of the T cells was assayed using a lymphocyte-tumor co-culture system in vitro. RESULTS: Ganoderic acid A enhanced tumor suppression of oxaliplatin in the xenograft model, while single administration showed no obvious anti-tumor effect. Ganoderic acid A didn't affect cell proliferation and apoptosis of HT-29 cells treated by oxaliplatin in vitro. Additionally, ganoderic acid A co-administered with oxaliplatin didn't impact T cell subtyping in the xenograft model. Cytotoxicity of T cells in co-administered mice was remarkably enhanced compared with oxaliplatin-treated mice. CONCLUSION: Our findings reveal that ganoderic acid A synergistically enhances tumor suppression of oxaliplatin possibly via increasing the cytotoxicity of T cells.


Asunto(s)
Antineoplásicos , Ácidos Heptanoicos , Triterpenos , Humanos , Ratones , Animales , Oxaliplatino/farmacología , Oxaliplatino/uso terapéutico , Linfocitos T , Ácidos Heptanoicos/farmacología , Apoptosis , Triterpenos/farmacología , Línea Celular Tumoral , Proliferación Celular , Antineoplásicos/uso terapéutico
5.
Int J Mol Sci ; 23(3)2022 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-35163524

RESUMEN

Anti-CD20 monoclonal antibodies (MAbs) have revolutionized the treatment of B-cell leukemia and lymphoma. However, many patients do not respond to such treatment due to either deficiency of the complementary immune response or resistance to apoptosis. Other currently available treatments are often inadequate or induce major side effects. Therefore, there is a constant need for improved therapies. The prostaglandin E2 receptor 4 (EP4) receptor has been identified as a promising therapeutic target for hematologic B-cell malignancies. Herein, we report that EP4 receptor agonists PgE1-OH and L-902688 have exhibited enhanced cytotoxicity when applied together with anti-CD20 MAbs rituximab, ofatumumab and obinutuzumab in vitro in Burkitt lymphoma cells Ramos, as well as in p53-deficient chronic lymphocytic leukemia (CLL) cells MEC-1. Moreover, the enhanced cytotoxic effects of EP4 receptor agonists and MAbs targeting CD20 have been identified ex vivo on primary lymphocytes B obtained from patients diagnosed with CLL. Incubation of cells with PgE1-OH and L-902688 preserved the expression of CD20 molecules, further confirming the anti-leukemic potential of EP4 receptor agonists in combination with anti-CD20 MAbs. Additionally, we demonstrated that the EP4 receptor agonist PgE-1-OH induced apoptosis and inhibited proliferation via the EP4 receptor triggering in CLL. This work has revealed very important findings leading towards the elucidation of the anticancer potential of PgE1-OH and L-902688, either alone or in combination with MAbs. This may contribute to the development of potential therapeutic alternatives for patients with B-cell malignancies.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígenos CD20/inmunología , Leucemia de Células B/metabolismo , Linfoma de Células B/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/agonistas , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ácidos Heptanoicos/farmacología , Humanos , Leucemia de Células B/tratamiento farmacológico , Linfoma de Células B/tratamiento farmacológico , Pirrolidinonas/farmacología , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Rituximab/farmacología , Rituximab/uso terapéutico , Tetrazoles/farmacología
6.
Exp Cell Res ; 412(1): 113025, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35026282

RESUMEN

Excessive oxidative stress and decreased antioxidant capacity of macrophages are initial factors which cause macrophages to transform to foam cells, which represents a key event in the progression of atherosclerosis (AS). BML-111, the analog of lipoxin A4 (LXA4) strongly attenuated high fat (HF) diet-induced atherosclerosis by activating NF-E2 related factor 2 (Nrf2). However, the effect was not through a specific LXA4 receptor (formyl peptide receptor 2, FPR2). BML-111 also strongly inhibited HF diet-induced promotion of MDA level, increased HDL level and decreased IL-1, MCP-1, IL-6, VCAM, ICAM and TNF-α level in aorta. In the in vitro experiments, LXA4 inhibited THP-1 cells to transform to foam cells via Nrf2 pathway. Our findings demonstrated that LXA4 and its analog prevented AS induced by HF diet in SD rats, under which the possible mechanism is through Keap1/Nrf2 pathway.


Asunto(s)
Aterosclerosis/metabolismo , Aterosclerosis/prevención & control , Ácidos Heptanoicos/farmacología , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Lipoxinas/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Aterosclerosis/etiología , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Células Espumosas/efectos de los fármacos , Células Espumosas/metabolismo , Células Espumosas/patología , Humanos , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de Lipoxina/metabolismo , Transducción de Señal/efectos de los fármacos , Células THP-1
7.
Int J Biol Macromol ; 189: 618-634, 2021 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-34437921

RESUMEN

Bud blight disease caused by groundnut bud necrosis virus (GBNV) is a serious constraint in the cultivation of agricultural crops such as legumes, tomato, chilies, potato, cotton etc. Owing to the significant damage caused by GBNV, an attempt was made to identify suitable organic antiviral agents through molecular modelling of the nucleocapsid Coat Protein of GBNV; molecular docking and molecular dynamics that disclosed the interaction of the ligands viz., Squalene and Ganoderic acid-A with coat protein of GBNV. Invitro inhibitory effect of Squalene and Ganoderic acid-A was examined in comparison with different concentrations, against GBNV in cowpea plants under glasshouse condition. The different concentrations of Squalene (50, 100, 150, 250 and 500 ppm) tested in vitro resulted in reduction of lesion numbers (1.69 cm2) as well as reduced virus titre in co-inoculation spray. The present study suggests the antiviral activity of Squalene by effectively fitting into binding site of coat protein of GBNV with favourable hydrophilic as well as strong hydrophobic interactions thereby challenging and blocking the binding of viral replication RNA with coat protein and propagation. The present organic antiviral molecules will be helpful in development of suitable eco-friendly formulations to mitigate GBNV infection disease in plants.


Asunto(s)
Antivirales/farmacología , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Simulación del Acoplamiento Molecular , Escualeno/farmacología , Tospovirus/química , Antivirales/química , Sitios de Unión , Fabaceae/virología , Ácidos Heptanoicos/química , Ácidos Heptanoicos/farmacología , Lanosterol/análogos & derivados , Lanosterol/química , Lanosterol/farmacología , Ligandos , Simulación de Dinámica Molecular , Escualeno/química
8.
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
9.
Biomed Pharmacother ; 141: 111909, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34328088

RESUMEN

Ganoderma lucidum is a legendary traditional Chinese medicine with various bioactivities. This study was conducted (a) to explore the in vitro fermentation of the water extracts of G. lucidum fruiting body with Lactobacillus acidophilus and Bifidobacterium breve and (b) to investigate the effect of fermentation broth (GLFB) on dexamethasone (DEX)-induced immunosuppressed mice. Our results demonstrated that probiotic fermentation of G. lucidum fruiting body extracts underwent structural changing of major ganoderic acid components, such as ganoderic acid A (GA) into GC2, and this fermentation process involves changing of several metabolic pathways in the probiotic strains. GLFB could significantly improve the immunity, intestinal integrity, and gut microbiota dysbiosis in DEX-treated mice, and the immunostimulatory activity of GLFB was found closely related to its direct regulation on the expansion of CD4+ T cells in Peyer's patches of mice. These data implied that probiotic fermentation of G. lucidum fruiting body extracts promoted its immunostimulatory activity via biotransformation of components such as GA. This research provides a theoretical support for the development and application of G. lucidum fermentation by probiotics.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Dexametasona/farmacología , Cuerpos Fructíferos de los Hongos/química , Inmunosupresores/farmacología , Probióticos/metabolismo , Reishi/metabolismo , Animales , Linfocitos T CD4-Positivos/metabolismo , Fermentación , Microbioma Gastrointestinal/efectos de los fármacos , Ácidos Heptanoicos/farmacología , Intestinos/efectos de los fármacos , Lanosterol/análogos & derivados , Lanosterol/farmacología , Recuento de Linfocitos , Masculino , Medicina Tradicional China , Ratones , Ratones Endogámicos BALB C , Ganglios Linfáticos Agregados/citología , Ganglios Linfáticos Agregados/efectos de los fármacos , Reishi/química
10.
Adv Clin Exp Med ; 30(10): 1031-1041, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34329545

RESUMEN

BACKGROUND: Atherosclerosis (AS), a chronic inflammatory disease, is a major contributor to deaths worldwide. Ganoderic acid A (GAA) has been widely applied for various diseases due to its excellent anti-inflammatory properties. OBJECTIVES: To investigate the underlying mechanism of GAA inhibition of inflammation and lipid deposition in human monocyte (THP-1) cells. MATERIAL AND METHODS: The Cell Counting Kit-8 (CCK-8) assay was used to assess the potential effect of GAA on the viability of THP-1 cells. The release of inflammatory cytokines and oxidative stress was measured using enzyme-linked immunosorbent assay (ELISA) and the corresponding kit, respectively. The levels of lipid deposition and total cholesterol (TC) were also evaluated. Next, the scavenger receptors and proteins in Notch1/PPARa/CD36 signaling were measured with western blot. As Notch1 was overexpressed in the THP-1 cells induced by oxidized low-density lipoprotein (ox-LDL), the above assays were performed again to confirm the underlying mechanism. RESULTS: Ganoderic acid A suppressed ox-LDL-induced inflammation and oxidative stress in THP-1 cells. At the same time, it inhibited the TC level and lipid deposition. The effects of GAA on alleviating inflammation, oxidative stress and lipid accumulation were relieved after the overexpression of Notch1 in the treated cells, and the effects of GAA on alleviating inflammation, oxidative stress and lipid accumulation were diminished. The PPARa activator also weakened the effects of GAA on relieving inflammation, oxidative stress and lipid accumulation in ox-LDL-induced THP-1 cells. CONCLUSIONS: Ganoderic acid A inhibits ox-LDL-induced macrophage inflammation and lipid deposition in THP-1 cells through Notch1/PPARa/CD36 signaling, which may provide theoretical guidance for the clinical applications of GAA in AS treatment.


Asunto(s)
Ácidos Heptanoicos/farmacología , Lipoproteínas LDL , Macrófagos/efectos de los fármacos , PPAR gamma , Antígenos CD36 , Humanos , Inflamación/prevención & control , Lanosterol/análogos & derivados , Receptor Notch1 , Transducción de Señal , Células THP-1
11.
Int J Mol Sci ; 22(11)2021 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-34074054

RESUMEN

Alzheimer's disease (AD) is thought to be caused by amyloid-ß (Aß) accumulation in the central nervous system due to deficient clearance. The aim of the present study was to investigate the effect of ganoderic acid A (GAA) on Aß clearance in microglia and its anti-AD activity. Aß degradation in BV2 microglial cells was determined using an intracellular Aß clearance assay. GAA stimulated autophagosome formation via the Axl receptor tyrosine kinase (Axl)/RAC/CDC42-activated kinase 1 (Pak1) pathway was determined by Western blot analyses, and fluorescence-labeled Aß42 was localized in lysosomes in confocal laser microscopy images. The in vivo anti-AD activity of GAA was evaluated by object recognition and Morris water maze (MWM) tests in an AD mouse model following intracerebroventricular injection of aggregated Aß42. The autophagy level in the hippocampus was assayed by immunohistochemical assessment against microtubule-associated proteins 1A/1B light-chain 3B (LC3B). Intracellular Aß42 levels were significantly reduced by GAA treatment in microglial cells. Additionally, GAA activated autophagy according to increased LC3B-II levels, with this increased autophagy stimulated by upregulating Axl and Pak1 phosphorylation. The effect of eliminating Aß by GAA through autophagy was reversed by R428, an Axl inhibitor, or IPA-3, a Pak1 inhibitor. Consistent with the cell-based assay, GAA ameliorated cognitive deficiency and reduced Aß42 levels in an AD mouse model. Furthermore, LC3B expression in the hippocampus was up-regulated by GAA treatment, with these GAA-specific effects abolished by R428. GAA promoted Aß clearance by enhancing autophagy via the Axl/Pak1 signaling pathway in microglial cells and ameliorated cognitive deficiency in an AD mouse model.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Autofagia/efectos de los fármacos , Ácidos Heptanoicos/farmacología , Lanosterol/análogos & derivados , Microglía/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Autofagosomas/efectos de los fármacos , Autofagosomas/metabolismo , Autofagia/genética , Línea Celular , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Inmunohistoquímica , Lanosterol/farmacología , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Microglía/patología , Prueba del Laberinto Acuático de Morris/efectos de los fármacos , Fosforilación , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Quinasas p21 Activadas/antagonistas & inhibidores , Quinasas p21 Activadas/metabolismo , Tirosina Quinasa del Receptor Axl
12.
Biosci Biotechnol Biochem ; 85(8): 1818-1829, 2021 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-33963744

RESUMEN

The effects of PADI4 and GAA on the senescence of Alzheimer's cells were explored in the present work. HT22 cells were treated with Aß25-35 to establish an Alzheimer's model and were then treated with different concentrations of GAA and transfected with a siPADI4 lentiviral vector. GAA could reverse the effects of Aß25-35 on inhibiting cell viability and promoting apoptosis and senescence. siPADI4 reduced Aß25-35-induced cell viability and upregulated Aß25-35-induced cell apoptosis and senescence, as well as partially reversed the effect of GAA on cells, and these results were confirmed by detecting the expressions of senescence- and apoptosis-related proteins. In addition, siPADI4 was found to promote the phosphorylation of Akt and mTOR, which was partially reversed by GAA. In conclusion, PADI4 mediates autophagy and participates in the role of GAA monomers in delaying the senescence of Alzheimer's cells through the Akt/mTOR pathway.


Asunto(s)
Enfermedad de Alzheimer/patología , Autofagia/fisiología , Senescencia Celular/efectos de los fármacos , Ácidos Heptanoicos/farmacología , Lanosterol/análogos & derivados , Arginina Deiminasa Proteína-Tipo 4/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/metabolismo , Línea Celular , Ácidos Heptanoicos/química , Humanos , Lanosterol/química , Lanosterol/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
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
14.
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
15.
Neurochem Res ; 46(7): 1725-1736, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33821438

RESUMEN

Neuroinflammation plays an important role in the onset and progression of neurodegenerative diseases. Microglia-mediated neuroinflammation have been proved to be the main reason for causing the neurodegenerative diseases. Ganoderic acid A (GAA), isolated from Ganoderma lucidum, showed anti-inflammatory effect in metabolism diseases. However, little research has been focused on the effect of GAA in neuroinflammation and the related mechanism. In the present study, lipopolysaccharide(LPS)-stimulated BV2 microglial cells were used to evaluate the anti-inflammatory capacity of GAA. Our data showed that GAA significantly suppressed LPS-induced BV2 microglial cells proliferation and activation in vitro. More strikingly, GAA promoted the conversion of BV2 microglial cells from M1 status induced by LPS to M2 status. Furthermore, GAA inhibited the pro-inflammatory cytokines release and promoted neurotrophic factor BDNF expression in LPS-induced BV2 microglial cells. Finally, we found that the expression of farnesoid-X-receptor (FXR) was prominently downregulated in LPS-stimulated BV2 microglial cells, antagonism of FXR with z-gugglesterone and FXR siRNA can reverse the effect of GAA in LPS-induced BV2 microglial cells. Taking together, our findings demonstrate that GAA can significantly inhibit LPS-induced neuroinflammation in BV2 microglial cells via activating FXR receptor.


Asunto(s)
Antiinflamatorios/farmacología , Ácidos Heptanoicos/farmacología , Inflamación/tratamiento farmacológico , Lanosterol/análogos & derivados , Microglía/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Inflamación/inducido químicamente , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Lanosterol/farmacología , Lipopolisacáridos , Ratones , Receptores Citoplasmáticos y Nucleares/genética , Factor de Necrosis Tumoral alfa/metabolismo
16.
Pflugers Arch ; 473(4): 683-695, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33474635

RESUMEN

The pro-resolving mechanism is a recently described endogenous process that controls inflammation. The present study evaluated components of this mechanism, including annexin 1 (ANXA1) and the formyl peptide receptor 2/ALX (FPR2/ALX) receptor, in the antihyperalgesic effect induced by electroacupuncture (EA) in an animal model of persistent peripheral inflammation. Male Swiss mice underwent intraplantar (i.pl.) injection with complete Freund's adjuvant (CFA). Mechanical hyperalgesia was assessed with von Frey monofilaments. Animals were treated with EA (2-10 Hz, ST36-SP6) or subcutaneous BML-111 injection (FPR2/ALX agonist) for 5 consecutive days. In a separate set of experiments, on the first and fifth days after CFA injection, animals received i.pl. WRW4 (FPR2/ALX antagonist) or naloxone (non-selective opioid receptor antagonist) before EA or BML-111 injection. Paw protein levels of FPR2/ALX and ANXA1 were evaluated on the second day after CFA injection by western blotting technique. EA and BML-111 reduced mechanical hyperalgesia. I.pl. naloxone or WRW4 prevented the antihyperalgesic effect induced by either EA or BML-111. EA increased ANXA1 but did not alter FPR2/ALX receptor levels in the paw. Furthermore, i.pl. pretreatment with WRW4 prevented the increase of ANXA1 levels induced by EA. This work demonstrates that the EA antihyperalgesic effect on inflammatory pain involves the ANXA1/FPR2/ALX pro-resolution pathway. This effect appears to be triggered by the activation of FPR2/ALX receptors and crosstalk communication with the opioid system.


Asunto(s)
Anexina A1/metabolismo , Electroacupuntura/métodos , Hiperalgesia/terapia , Dolor Nociceptivo/terapia , Receptores de Formil Péptido/metabolismo , Receptores Opioides/metabolismo , Animales , Adyuvante de Freund/toxicidad , Ácidos Heptanoicos/farmacología , Hiperalgesia/etiología , Hiperalgesia/metabolismo , Masculino , Ratones , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Nocicepción/efectos de los fármacos , Dolor Nociceptivo/etiología , Dolor Nociceptivo/metabolismo , Receptores de Formil Péptido/antagonistas & inhibidores , Receptores Opioides/uso terapéutico
17.
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
18.
Immunol Lett ; 230: 27-35, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33347917

RESUMEN

Tumor angiogenesis plays a vital role in carcinogenesis, cancer progression, and metastasis. Lipoxin A4 (LXA4) is an endogenously-produced family of effective anti-inflammatory with a potent inhibitory effect on angiogenesis. However, BML-111, a LXA4 agonist, its governing tumor-derived endothelial cells (Td-EC) mechanisms remain unknown. In the present study, we utilized VEGF or CoCl2 to mimic tumor microenvironment in vitro to study the effect of BML-111 on angiogenesis and permeability of Td-EC, and preliminarily explore its specific mechanism. Data suggested that BML-111 inhibited viability, migration and angiogenesis in VEGF or CoCl2-treated Td-EC by modulating MMP2/9-TIMP1, and decreasing the production of HIF-1α and COX-2 level. In addition, we observed that BML-111 inhibited Td-EC permeability induced by VEGF or CoCl2, through the stabilization of VE-cadherin/ß-catenin-dependent adherens junctions and TRPC1 pathway. Nevertheless, these effects could be blocked by BOC-2 which was the specific inhibitor of FPR2/ALX (the receptor of LXA4).These results suggest that BML-111 may have inhibitory effects on VEGF or CoCl2-induced migration, angiogenesis and permeability in tumor-derived endothelial cells.


Asunto(s)
Moduladores de la Angiogénesis/farmacología , Endotelio Vascular/fisiología , Ácidos Heptanoicos/farmacología , Lipoxinas/agonistas , Neoplasias/tratamiento farmacológico , Uniones Adherentes/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Cobalto/metabolismo , Células Hep G2 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neoplasias/irrigación sanguínea , Neoplasias/patología , Neovascularización Patológica , Transducción de Señal , Canales Catiónicos TRPC/metabolismo , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
Chin J Nat Med ; 18(12): 934-940, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33357724

RESUMEN

A contributory role of oxidative stress and protection by antioxidant nutrients have been suspected in cataract formation. Ganoderic acid A (GAA), an effective lanostane triterpene, is widely reported as an antioxidant. The aim of this study is to investigate the potential effects of GAA on cataract formation. After lens epithelial cells (LECs) were exposed to UVB radiation for different periods, cell viability, apoptosis-related protein levels, malondialdehyde (MDA) and superoxide dismutase (SOD) activities were monitored. We found that cell viability, the Bcl-2/Bax ratio and SOD activity were increased, while Cleaved caspase-3 levels and MDA activity were decreased compared with those in UVB-impaired LECs after GAA treated. Furthermore, GAA activated PI3K/AKT in UVB-impaired LECs and effectively delayed the occurrence of lens opacity in vitro. In conclusion, these findings demonstrated that GAA exhibited protective functions in SRA01/04 cells and rat lenses against UVB-evoked impairment through elevating cell viability and antioxidant activity, inhibiting cell apoptosis, activating the PI3K/AKT pathway and delaying lens opacity.


Asunto(s)
Catarata/prevención & control , Células Epiteliales/efectos de los fármacos , Ácidos Heptanoicos/farmacología , Lanosterol/análogos & derivados , Cristalino/citología , Rayos Ultravioleta/efectos adversos , Animales , Apoptosis , Línea Celular , Supervivencia Celular , Células Epiteliales/efectos de la radiación , Humanos , Lanosterol/farmacología , Cristalino/efectos de la radiación , Malondialdehído/metabolismo , Ratas , Superóxido Dismutasa/metabolismo
20.
Molecules ; 25(12)2020 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-32604968

RESUMEN

Excessive exposure to UV, especially UVB, is the most important risk factor for skin cancer and premature skin aging. The identification of the specialized pro-resolving lipid mediators (SPMs) challenged the preexisting paradigm of how inflammation ends. Rather than a passive process, the resolution of inflammation relies on the active production of SPMs, such as Lipoxins (Lx), Maresins, protectins, and Resolvins. LXA4 is an SPM that exerts its action through ALX/FPR2 receptor. Stable ALX/FPR2 agonists are required because SPMs can be quickly metabolized within tissues near the site of formation. BML-111 is a commercially available synthetic ALX/FPR2 receptor agonist with analgesic, antioxidant, and anti-inflammatory properties. Based on that, we aimed to determine the effect of BML-111 in a model of UVB-induced skin inflammation in hairless mice. We demonstrated that BML-111 ameliorates the signs of UVB-induced skin inflammation by reducing neutrophil recruitment and mast cell activation. Reduction of these cells by BML-111 led to lower number of sunburn cells formation, decrease in epidermal thickness, collagen degradation, cytokine production (TNF-α, IL-1ß, IL-6, TGF, and IL-10), and oxidative stress (observed by an increase in total antioxidant capacity and Nrf2 signaling pathway), indicating that BML-111 might be a promising drug to treat skin disorders.


Asunto(s)
Dermatitis/prevención & control , Ácidos Heptanoicos/administración & dosificación , Protectores contra Radiación/administración & dosificación , Receptores de Lipoxina/antagonistas & inhibidores , Animales , Antígenos CD59/metabolismo , Dermatitis/etiología , Dermatitis/metabolismo , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/metabolismo , Relación Dosis-Respuesta a Droga , Ácidos Heptanoicos/farmacología , Lipoxinas/metabolismo , Ratones , Ratones Pelados , Protectores contra Radiación/farmacología , Rayos Ultravioleta/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...