Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.788
Filter
2.
J Pharm Pharmacol ; 76(4): 354-367, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38330446

ABSTRACT

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.


Subject(s)
Antineoplastic Agents , Autophagy , Carcinoma, Non-Small-Cell Lung , Heptanoic Acids , Lanosterol , Lung Neoplasms , MicroRNAs , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Cell Proliferation , Cisplatin/pharmacology , Cytochrome P-450 CYP1A1/drug effects , Cytochrome P-450 CYP1A1/metabolism , Drug Resistance, Neoplasm , Heptanoic Acids/pharmacology , Heptanoic Acids/therapeutic use , Lanosterol/analogs & derivatives , Lanosterol/pharmacology , Lanosterol/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MicroRNAs/drug effects , MicroRNAs/metabolism , RNA, Circular/drug effects , RNA, Circular/metabolism , X-ray Repair Cross Complementing Protein 1/drug effects , X-ray Repair Cross Complementing Protein 1/metabolism
3.
Aging (Albany NY) ; 15(23): 13669-13679, 2023 Nov 17.
Article in English | MEDLINE | ID: mdl-38048213

ABSTRACT

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.


Subject(s)
Heptanoic Acids , Hypercholesterolemia , Hyperlipidemias , Humans , Mice , Animals , Atorvastatin/pharmacology , Atorvastatin/therapeutic use , Hypercholesterolemia/complications , Hypercholesterolemia/drug therapy , Monocytes/metabolism , Heptanoic Acids/pharmacology , Heptanoic Acids/therapeutic use , Pyrroles/pharmacology , Hyperlipidemias/drug therapy , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Brain/metabolism
4.
Adv Med Sci ; 68(2): 258-264, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37478516

ABSTRACT

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.


Subject(s)
Carcinoma, Hepatocellular , Heptanoic Acids , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Liver Neoplasms , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Rosuvastatin Calcium/pharmacology , Atorvastatin/pharmacology , Simvastatin/pharmacology , Simvastatin/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Reactive Oxygen Species , Heptanoic Acids/therapeutic use , Pyrroles/therapeutic use , Fluorobenzenes/therapeutic use , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Liver Neoplasms/drug therapy , Glucose
5.
Pharmacol Res Perspect ; 11(3): e01089, 2023 06.
Article in English | MEDLINE | ID: mdl-37186070

ABSTRACT

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.


Subject(s)
Anticholesteremic Agents , Coronary Disease , Heptanoic Acids , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Humans , Atorvastatin/therapeutic use , Cholesterol, LDL , Anticholesteremic Agents/therapeutic use , Anticholesteremic Agents/pharmacology , Heptanoic Acids/pharmacology , Heptanoic Acids/therapeutic use , Pyrroles , Triglycerides , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Coronary Disease/drug therapy , Coronary Disease/chemically induced
6.
Clin Sci (Lond) ; 135(16): 1945-1980, 2021 08 27.
Article in English | MEDLINE | ID: mdl-34401905

ABSTRACT

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.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Asthma/drug therapy , Receptors, Eicosanoid/agonists , Receptors, Eicosanoid/antagonists & inhibitors , Acetates/pharmacology , Acetates/therapeutic use , Animals , Anti-Asthmatic Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Asthma/metabolism , Asthma/physiopathology , Benzoquinones/pharmacology , Benzoquinones/therapeutic use , Biomarkers/metabolism , Cyclopropanes/pharmacology , Cyclopropanes/therapeutic use , Heptanoic Acids/pharmacology , Heptanoic Acids/therapeutic use , Humans , Lung/drug effects , Lung/metabolism , Lung/physiopathology , Mice , Quinolines/pharmacology , Quinolines/therapeutic use , Receptors, Eicosanoid/drug effects , Sulfides/pharmacology , Sulfides/therapeutic use
7.
Chem Biol Interact ; 344: 109529, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-34029542

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Fibrosis/drug therapy , Heptanoic Acids/therapeutic use , Kidney Diseases/drug therapy , Lanosterol/analogs & derivatives , Signal Transduction/drug effects , Animals , Carbon Tetrachloride , Fibrosis/chemically induced , Fibrosis/pathology , Janus Kinase 2/metabolism , Kidney Diseases/chemically induced , Kidney Diseases/pathology , Lanosterol/therapeutic use , Male , Mice, Inbred ICR , Oxidative Stress/drug effects , Smad3 Protein/metabolism , Thioredoxin-Disulfide Reductase/metabolism , Thioredoxins/metabolism , Transforming Growth Factor beta1/metabolism , rho-Associated Kinases/metabolism
8.
Inflammation ; 44(5): 1908-1915, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34037898

ABSTRACT

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.


Subject(s)
Asthma/chemically induced , Asthma/drug therapy , Heptanoic Acids/therapeutic use , Inflammation Mediators/antagonists & inhibitors , Lanosterol/analogs & derivatives , Ovalbumin/toxicity , Animals , Asthma/metabolism , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Dose-Response Relationship, Drug , Female , Heptanoic Acids/pharmacology , Inflammation Mediators/metabolism , Lanosterol/pharmacology , Lanosterol/therapeutic use , Mice , Mice, Inbred BALB C , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/metabolism
9.
Biochem Pharmacol ; 188: 114561, 2021 06.
Article in English | MEDLINE | ID: mdl-33857491

ABSTRACT

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.


Subject(s)
Antidepressive Agents/therapeutic use , Depression/metabolism , Heptanoic Acids/therapeutic use , Lanosterol/analogs & derivatives , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Receptors, Cytoplasmic and Nuclear/deficiency , Synapses/metabolism , Animals , Antidepressive Agents/pharmacology , Depression/drug therapy , Depression/psychology , Heptanoic Acids/pharmacology , Lanosterol/pharmacology , Lanosterol/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Social Defeat , Synapses/drug effects , Synapses/genetics
10.
Biochem Pharmacol ; 185: 114422, 2021 03.
Article in English | MEDLINE | ID: mdl-33482151

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Heptanoic Acids/therapeutic use , Lanosterol/analogs & derivatives , Multiple Sclerosis/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Regeneration/physiology , Remyelination/physiology , Animals , Anti-Inflammatory Agents/pharmacology , Female , Heptanoic Acids/pharmacology , Lanosterol/pharmacology , Lanosterol/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Myelin Sheath/pathology , Regeneration/drug effects , Remyelination/drug effects
11.
Immunol Lett ; 226: 1-6, 2020 10.
Article in English | MEDLINE | ID: mdl-32565114

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Heptanoic Acids/therapeutic use , Lanosterol/analogs & derivatives , Synoviocytes/pathology , Animals , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Humans , Janus Kinase 3/metabolism , Lanosterol/therapeutic use , Male , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Signal Transduction
12.
Int Immunopharmacol ; 85: 106625, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32485356

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Heptanoic Acids/pharmacology , Protein Serine-Threonine Kinases/metabolism , Triple Negative Breast Neoplasms/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Female , Glycogen Synthase Kinase 3 beta/metabolism , Heptanoic Acids/therapeutic use , Humans , Interleukin-8/metabolism , Lung/pathology , Macrophages/drug effects , Mice, Inbred BALB C , Neoplasm Metastasis/prevention & control , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment , Tumor-Associated Macrophages , Xenograft Model Antitumor Assays
13.
Int Immunopharmacol ; 84: 106543, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32353688

ABSTRACT

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.


Subject(s)
Heptanoic Acids/therapeutic use , Janus Kinase 2/metabolism , Lanosterol/analogs & derivatives , Myocardial Reperfusion Injury/drug therapy , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cytokines/blood , Heptanoic Acids/pharmacology , Lanosterol/pharmacology , Lanosterol/therapeutic use , Male , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Myocardium/pathology , Rats, Sprague-Dawley , Signal Transduction/drug effects , Ventricular Function, Left/drug effects
14.
Pharmacology ; 105(9-10): 568-575, 2020.
Article in English | MEDLINE | ID: mdl-31940650

ABSTRACT

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.


Subject(s)
Heptanoic Acids/pharmacology , Lanosterol/analogs & derivatives , Lung/drug effects , Plant Extracts/pharmacology , Protective Agents/pharmacology , Pulmonary Fibrosis/drug therapy , Animals , Bleomycin/toxicity , Cytokines/blood , Heptanoic Acids/therapeutic use , Lanosterol/pharmacology , Lanosterol/therapeutic use , Lung/metabolism , Lung/pathology , Male , Malondialdehyde/blood , Mice , Mice, Inbred ICR , NF-kappa B/metabolism , Peroxidase/metabolism , Phytotherapy , Plant Extracts/administration & dosage , Plant Extracts/therapeutic use , Protective Agents/therapeutic use , Pulmonary Edema/drug therapy , Pulmonary Fibrosis/chemically induced , Smad3 Protein/metabolism , Superoxide Dismutase/blood , Transforming Growth Factor beta/metabolism
15.
J Diabetes Complications ; 34(3): 107480, 2020 03.
Article in English | MEDLINE | ID: mdl-31902653

ABSTRACT

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.


Subject(s)
Amlodipine/therapeutic use , Behavior Therapy/methods , Heptanoic Acids/therapeutic use , Metabolic Syndrome/therapy , Pyrroles/therapeutic use , Adult , Aged , Amlodipine/administration & dosage , Atorvastatin/administration & dosage , Biomarkers/blood , Blood Pressure/physiology , Cardiometabolic Risk Factors , Combined Modality Therapy , Drug Combinations , Female , Humans , Life Style , Lipids/blood , Lipoproteins, LDL/blood , Male , Metabolic Syndrome/blood , Metabolic Syndrome/physiopathology , Middle Aged , Oxidative Stress/physiology , Placebos , Risk Reduction Behavior
16.
Int Immunopharmacol ; 79: 106108, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31881376

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Diabetic Nephropathies/drug therapy , Heptanoic Acids/therapeutic use , Inflammation/drug therapy , Mesangial Cells/metabolism , NF-E2-Related Factor 2/metabolism , Animals , Cell Line , Extracellular Matrix/drug effects , Glucose/metabolism , Humans , Mesangial Cells/pathology , Molecular Targeted Therapy , Oxidative Stress/drug effects , Rats , Signal Transduction
17.
Biomed Pharmacother ; 112: 108595, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30784911

ABSTRACT

This study was designed to investigate the inflammatory responses in fat embolism syndrome (FES) and the relationship of ALX/FPR2 receptors and lipoxin A4 (LXA4) in FES models. In this model, lung injury score, lung tissue wet-to-dry (W/D) ratio and total protein concentration in bronchoalveolar lavage fluid (BALF) were increased compared with those of the control group. Meanwhile, the number of leukocytes and neutrophils was significantly increased in the FES group, as was the myeloperoxidase (MPO) activity and mRNA expression. In addition, the release of TNF-α and IL-1ß was increased. Then, we explored whether LXA4 and ALX/FPR2 were involved in the pathological process of FES. The LXA4 concentration in the experimental groups was markedly higher than that in the control group. At the same time, the protein and mRNA levels of ALX/FPR2 were upregulated in the rat model of FES. Moreover, rats treated with BML-111, an agonist for the ALX/FPR2 receptor of LXA4, showed a lower inflammatory response than mice treated with fat alone. However, the role of BML-111 in fat emboli (FE)-induced acute lung injury (ALI) was attenuated by BOC-2, an antagonist of the ALX/FPR2 receptor of LXA4. Our results demonstrated that the inflammatory response may play an important role in the pathogenesis of FES and that the activation of the ALX/FPR2 receptor for LXA4 can decrease the inflammatory response and may be a therapeutic target for FE-induced ALI.


Subject(s)
Embolism, Fat/metabolism , Embolism, Fat/pathology , Lung Injury/metabolism , Lung Injury/pathology , Receptors, Lipoxin/metabolism , Animals , Embolism, Fat/drug therapy , Heptanoic Acids/pharmacology , Heptanoic Acids/therapeutic use , Lung Injury/drug therapy , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Lipoxin/agonists
18.
Int Immunopharmacol ; 69: 289-298, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30753968

ABSTRACT

The timely resolution of pulmonary inflammation coordinated by endogenous pro-resolving mediators helps limit lung tissue injury, but few endogenous pro-resolving mediators that are normally operative during acute inflammation. The protective effects of BML-111 (5(S)-6(R)-7-trihydroxyheptanoic acid methyl ester), a potent commercially available anti-inflammatory and pro-resolving mediator, on ventilation-induced lung injury (VILI) have been extensively studied, but its characteristics as a pro-resolving mediator have not. Here, anesthetized Sprague-Dawley rats were ventilated with a high tidal volume (20 mL/kg, HVT) for 1 h and randomly allocated to recover for 6, 12, 24, 48, 72, 96 or 168 h; BML-111 was administered at the peak of inflammation to evaluate its pro-resolving effect on VILI. The one-hour HVT induced a maximal pulmonary inflammatory response at 12 h that was largely resolved by 72 h. BML-111 largely resolved the maximal inflammatory response at 48 h; the resolution interval (Ri) was shortened by 26 h. Similarly, HVT elicited a time course of changes in histopathology and pulmonary edema, and BML-111 alleviates these changes. Mechanistically, neutrophil apoptosis was significantly increased in BML-111-treated rats subjected to HVT. The apoptosis inhibitor z-VAD-fmk partially reversed the proapoptotic actions of BML-111 on neutrophil and the resolving effects of BML-111 on VILI but had no effect alone. Importantly, the HVT treatment activated the nuclear factor E2-related factor 2(Nrf2)/heme oxygenase-1(HO-1) and NF-κB signaling pathways in the lung tissue, and BML-111 further induced Nrf2 and HO-1 expression but inhibited the NF-κB pathway. Intriguingly, when we inhibited the Nrf2/HO-1 pathway with the HO-1 inhibitor zinc protoporphyrin IX (ZnPPIX), Nrf2 expression was further increased, but the inhibitory effects of BML-111 on the NF-κB pathway and on the subsequent inflammatory response, and the proapoptotic actions on neutrophil were reversed. The results suggest that BML-111 promotes the resolution of HVT-induced inflammation to mitigate VILI in rats, perhaps by modulating the Nrf2/HO-1 and NF-κB pathways and subsequently increasing neutrophil apoptosis.


Subject(s)
Heptanoic Acids/therapeutic use , Inflammation/drug therapy , Lung/drug effects , Neutrophils/pathology , Ventilator-Induced Lung Injury/drug therapy , Animals , Apoptosis , Cells, Cultured , Disease Models, Animal , Heme Oxygenase-1/metabolism , Humans , Lung/pathology , Male , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Protoporphyrins/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction
19.
Respir Res ; 19(1): 243, 2018 Dec 05.
Article in English | MEDLINE | ID: mdl-30518355

ABSTRACT

BACKGROUND: Acute lung injury (ALI) is a life-threatening lung disease where alveolar macrophages (AMs) play a central role both in the early phase to initiate inflammatory responses and in the late phase to promote tissue repair. In this study, we examined whether BML-111, a lipoxin A4 receptor agonist, could alter the phenotypes of AM and thus present prophylactic benefits for ALI. METHODS: In vitro, isolated AMs were treated with lipopolysaccharide (LPS) to induce ALI. In response to BML-111 pre-treatment, apoptosis and autophagy of AMs were examined by flow cytometry, and by measuring biomarkers for each process. The potential involvement of MAPK1 and mTOR signaling pathway was analyzed. In vivo, an LPS-induced septic ALI model was established in rats and the preventative significance of BML-111 was assessed. On the cellular and molecular levels, the pro-inflammatory cytokines TNF-α and IL-6 from bronchoalveolar lavage were measured by ELISA, and the autophagy in AMs examined using Western blot. RESULTS: BML-111 inhibited apoptosis and induced autophagy of AMs in response to ALI inducer, LPS. The enhancement of autophagy was mediated through the suppression of MAPK1 and MAPK8 signaling, but independent of mTOR signaling. In vivo, BML-111 pre-treatment significantly alleviated LPS-induced ALI, which was associated with the reduction of apoptosis, the dampened production of pro-inflammatory cytokines in the lung tissue, as well as the increase of autophagy of AMs. CONCLUSIONS: This study reveals the prophylactic significance of BML-111 in ALI and the underlying mechanism: by targeting the MAPK signaling but not mTOR pathway, BML-111 stimulates autophagy in AMs, attenuates the LPS-induced cell apoptosis, and promotes the resolution of ALI.


Subject(s)
Acute Lung Injury/enzymology , Autophagy/physiology , Heptanoic Acids/therapeutic use , MAP Kinase Signaling System/physiology , Macrophages, Alveolar/enzymology , Receptors, Lipoxin/agonists , Acute Lung Injury/chemically induced , Acute Lung Injury/prevention & control , Animals , Autophagy/drug effects , Enzyme Activation/drug effects , Enzyme Activation/physiology , Heptanoic Acids/pharmacology , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , Macrophages, Alveolar/drug effects , Male , Rats , Rats, Sprague-Dawley
20.
Chem Biol Interact ; 290: 77-87, 2018 Jun 25.
Article in English | MEDLINE | ID: mdl-29852127

ABSTRACT

Obesity and its major co-morbidity, type 2 diabetes, have been an alarming epidemic prevalence without an effective treatment available. Sterol regulatory element-binding proteins (SREBPs) are major transcription factors regulating the expression of genes involved in biosynthesis of cholesterol, fatty acid and triglyceride. Therefore, inhibition of SREBP pathway may be a useful strategy to treat obesity with type 2 diabetes. Here, we identify a small molecule, Ganoderic Acid A (GAA), inhibits the SREBP expression and decreases the cellular levels of cholesterol and fatty acid in vitro. GAA also ameliorates body weight gain and fat accumulation in liver or adipose tissues, and improves serum lipid levels and insulin sensitivity in high fat diet (HFD)-induced obese mice. Consistently, GAA regulates SREBPs target genes and metabolism associated genes in liver or adipose tissues, which may directly contribute to the lower lipid level and improvement of insulin resistance. Taken together, GAA could be a potential leading compound for development of drugs for the prevention of obesity and insulin resistance.


Subject(s)
Diet, High-Fat , Heptanoic Acids/pharmacology , Lanosterol/analogs & derivatives , Lipid Metabolism/drug effects , Obesity/etiology , Sterol Regulatory Element Binding Proteins/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Aspartate Aminotransferases/analysis , Aspartate Aminotransferases/blood , Body Weight/drug effects , Cholesterol/blood , Energy Metabolism/drug effects , Glucose Tolerance Test , Hep G2 Cells , Heptanoic Acids/chemistry , Heptanoic Acids/therapeutic use , Humans , Insulin Resistance , Lanosterol/chemistry , Lanosterol/pharmacology , Lanosterol/therapeutic use , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Obesity/prevention & control , Signal Transduction/drug effects , Triglycerides/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...