Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters











Publication year range
1.
Int J Mol Med ; 53(4)2024 Apr.
Article in English | MEDLINE | ID: mdl-38362920

ABSTRACT

Due to molecular forces, biomacromolecules assemble into liquid condensates or solid aggregates, and their corresponding formation and dissolution processes are controlled. Protein homeostasis is disrupted by increasing age or environmental stress, leading to irreversible protein aggregation. Hypoxic pressure is an important factor in this process, and uncontrolled protein aggregation has been widely observed in hypoxia­related conditions such as neurodegenerative disease, cardiovascular disease, hypoxic brain injury and cancer. Biomolecular condensates are also high­order complexes assembled from macromolecules. Although they exist in different phase from protein aggregates, they are in dynamic balance under certain conditions, and their activation or assembly are considered as important regulatory processes in cell survival with hypoxic pressure. Therefore, a better understanding of the relationship between hypoxic stress, protein aggregation and biomolecular condensation will bring marked benefits in the clinical treatment of various diseases. The aim of the present review was to summarize the underlying mechanisms of aggregate assembly and dissolution induced by hypoxic conditions, and address recent breakthroughs in understanding the role of aggregates in hypoxic­related diseases, given the hypotheses that hypoxia induces macromolecular assemblage changes from a liquid to a solid phase, and that adenosine triphosphate depletion and ATP­driven inactivation of multiple protein chaperones play important roles among the process. Moreover, it is anticipated that an improved understanding of the adaptation in hypoxic environments could extend the overall survival of patients and provide new strategies for hypoxic­related diseases.


Subject(s)
Cardiovascular Diseases , Neurodegenerative Diseases , Humans , Protein Aggregates , Hypoxia , Adenosine Triphosphate
2.
Environ Toxicol ; 39(5): 2642-2654, 2024 May.
Article in English | MEDLINE | ID: mdl-38214030

ABSTRACT

BACKGROUND: The exact cause of intracranial aneurysms (IA) is still unclear. However, pro-inflammatory factors are known to contribute to IA progression. The specific changes in the immune microenvironment of IAs remain largely unexplored. METHODS: This study analyzed single-cell sequencing data from a male mouse model of brain aneurysm, focusing on samples before and after elastase-induced Willis aneurysms. The data helped identify eight distinct cell subpopulations: fibroblasts, macrophages, NK cells, endothelial cells, B cells, granulocytes, and monocytes. The study also involved bulk RNA sequencing of 97 IA samples, utilizing ssGSEA and CIBERSORT algorithms for analysis. Intercellular communication among these cells was inferred to understand the immune dynamics in IA. RESULTS: The study found that fibroblasts and macrophages are predominant in various disease states of IA. Notably, the onset of IA was marked by a significant increase in fibroblasts and a decrease in macrophages. There was a marked increase in cellular interactions, especially involving macrophages, at the onset of the disease. Through enrichment analysis, 12 potential immunogenic biomarkers were identified. Of these, Rgs1 emerged as a critical molecule in IA formation, confirmed through secondary validation in a single-cell sequencing dataset. CONCLUSION: This comprehensive analysis of immune cell composition and intercellular communication in IA tissues highlights the significant roles of macrophages and the molecule Rgs1. These findings shed light on the physiological and pathological conditions of IA, offering new insights into its immune microenvironment.


Subject(s)
Intracranial Aneurysm , Mice , Animals , Male , Intracranial Aneurysm/genetics , Intracranial Aneurysm/pathology , Endothelial Cells/pathology , Multiomics , Disease Models, Animal , Biomarkers
3.
Biomed Pharmacother ; 165: 115113, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37418974

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is one of the most common liver diseases with limited treatment options. Moreover, its prevalence is doubled in type 2 diabetes mellitus (T2DM). Kaempferol (KAP) is a flavonoid compound that has been suggested to have beneficial effects on NAFLD, but studies on the mechanism are lacking, especially in the diabetic state. Herein, we investigated the effect of KAP on NAFLD associated with T2DM and its underlying mechanism in vitro and in vivo. The results of in vitro studies indicated that KAP treatment (10-8-10-6 M) significantly reduced lipid accumulation in oleic acid-induced HepG2 cells. Moreover, in the T2DM animal model of db/db mice, we confirmed that KAP (50 mg/kg) significantly reduced lipid accumulation and improved liver injury. Mechanistic studies in vitro and in vivo showed that Sirtuin 1 (Sirt1)/AMP-activated protein kinase (AMPK) signal was involved in KAP regulation of hepatic lipid accumulation. KAP treatment activated Sirt1 and AMPK, upregulated the levels of fatty acid oxidation-related protein proliferator activated receptor gamma coactivator 1α (PGC1α); and downregulated lipid synthesis-related proteins, including acetyl-coA carboxylase (ACC), fatty acid synthase (FASN), and sterol regulatory element-binding protein 1 (SREBP1). Furthermore, the curative effect of KAP on lipid accumulation was abolished by siRNA-mediated knockdown of either Sirt1 or AMPK. Collectively, these findings suggest that KAP may be a potential therapeutic agent for NAFLD associated with T2DM by regulating hepatic lipid accumulation through activation of Sirt1/AMPK signaling.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Non-alcoholic Fatty Liver Disease , Mice , Animals , Humans , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Sirtuin 1/metabolism , AMP-Activated Protein Kinases/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Kaempferols/pharmacology , Kaempferols/therapeutic use , Liver , Signal Transduction , Lipid Metabolism , Hep G2 Cells , Lipids/pharmacology , Mice, Inbred C57BL
4.
Biomed Res Int ; 2022: 4013707, 2022.
Article in English | MEDLINE | ID: mdl-36110124

ABSTRACT

The edaravone and dexborneol concentrated solution for injection (edaravone-dexborneol) is a medication used clinically to treat neurological impairment induced by ischemic stroke. This study was aimed at investigating the preventive effects and the underlying mechanisms of edaravone-dexborneol on cerebral ischemic injury. A rat four-vessel occlusion (4-VO) model was established, and the neuronal injury and consequent neurological impairment of rats was investigated. Brain tissue malondialdehyde (MDA), myeloperoxidase (MPO), and nitric oxide (NO) levels were determined. The levels of proteins in mitogen-activated protein kinases (MAPKs), nuclear factor erythroid 2-related factor 2 (Nrf2), and nuclear factor-κB (NF-κB) signaling pathways were determined by western immunoblotting. The function of mitogen-activated protein kinase phosphatase 1 (MKP-1) was investigated using both western blot and immunofluorescence methods, and the effect of the MKP-1 inhibitor, (2E)-2-benzylidene-3-(cyclohexylamino)-3H-inden-1-one (BCI), was investigated. The results indicated that edaravone-dexborneol alleviated neurological deficiency symptoms and decreased apoptosis and neuron damage in the hippocampal CA1 area of the ischemic rats. Edaravone-dexborneol increased the MKP-1 level; decreased the phosphorylation of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase (p38 MAPK); inhibited NF-κB p65 activation; and boosted Nrf2 activation, all of which were partially reversed by the MKP-1 inhibitor, BCI. The above results indicated that the upregulation of MKP-1 contributed to the protective effects of edaravone-dexborneol against ischemic brain injury. Our findings support the hypothesis that edaravone-dexborneol can alleviate cerebral ischemic injury via the upregulation of MKP-1, which inhibits MAPKs and activates Nrf2.


Subject(s)
Brain Injuries , Dual Specificity Phosphatase 1 , Edaravone , NF-kappa B , Animals , Dual Specificity Phosphatase 1/metabolism , Edaravone/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Malondialdehyde , NF-E2-Related Factor 2 , Nitric Oxide , Peroxidase , Rats , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Pharmacol Res ; 175: 105989, 2022 01.
Article in English | MEDLINE | ID: mdl-34800628

ABSTRACT

Chronic cerebral ischemia (CCI) refers to long-term hypoperfusion of cerebral blood flow with the main clinical manifestations of progressive cognitive impairment. The pathological mechanism of CCI is complex, and there is a lack of effective treatments. Salvianolic acid A (SalA) is a neuroprotective extract of Salvia miltiorrhiza with the effects of anti-inflammation and anti-apoptosis. In this study, the effect of SalA on cognitive function and Drd2/Cryab/NF-κB signaling pathway in rats with CCI was investigated. Morris water maze and open field test were used to observe the effects of SalA on the cognitive function of CCI rats. The pathological changes in the brain were observed by HE, Nissl, and LFB staining. TUNEL staining, enzyme-linked immunosorbent assay, and western blot analysis were used to detect the inflammatory and apoptosis in the cortex and hippocampus. The expression of Drd2/Cryab/NF-κB pathway-related molecules and Drd2 localization were detected by western blotting and dual immunofluorescence, respectively. SH-SY5Y cells were exposed to chronic hypoglycemic and hypoxic injury in vitro, and Drd2 inhibitor haloperidol was used to verify the involved pathway. The results showed that SalA could improve the cognitive function of CCI rats, reduce pathological damage of cortex and hippocampus, inhibit neuroinflammation and apoptosis, and suppress the activation of NF-κB by regulating Drd2/Cryab pathway. And SalA inhibited NF-κB activation and nuclear translocation in SH-SY5Y cells by upregulating Drd2/Cryab pathway, which was reversed by haloperidol interference. In conclusion, SalA could relieve CCI-induced cognitive impairment in rats, at least partly through the Drd2/Cryab/NF-κB pathway.


Subject(s)
Brain Ischemia/drug therapy , Caffeic Acids/therapeutic use , Cognitive Dysfunction/drug therapy , Lactates/therapeutic use , Neuroinflammatory Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Caffeic Acids/pharmacology , Cell Hypoxia/drug effects , Cell Line, Tumor , Chronic Disease , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Crystallins/metabolism , Glucose/metabolism , Humans , Lactates/pharmacology , Male , Microtubule-Associated Proteins/metabolism , NF-kappa B/metabolism , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Neuroprotective Agents/pharmacology , Rats, Wistar , Receptors, Dopamine D2/metabolism
6.
Molecules ; 26(9)2021 May 03.
Article in English | MEDLINE | ID: mdl-34063645

ABSTRACT

Drug combinations have been the hotspot of the pharmaceutical industry, but the promising applications are limited by the unmet solubility and low bioavailability. In this work, novel cocrystals, consisting of two antithrombotic drugs with poor solubility and low bioavailability in vivo, namely, apixaban (Apx) and quercetin (Que), were developed to discover a potential method to improve the poor solubility and internal absorption of the drug combination. Compared with Apx, the dissolution behavior of Apx-Que (1:1) and Apx-Que-2ACN (1:1:2) was enhanced significantly, while the physical mixture of the chemicals failed to exhibit the advantages. The dissolution improvements of Apx-Que-2ACN could be explained by the fact that the solid dispersion-like structure and column-shaped cage of Que accelerated the access of the solvent to the inner layer of Apx. The fracture of the hydrogen bonds of Apx, which was the joint of the adjacent Que chains, facilitated the break-up of the structures. Besides, the bioavailability of Apx-Que was increased compared with the physical mixture and Apx, and Apx-Que remained stable in high temperature and illumination conditions. Therefore, a drug-drug cocrystal of two antithrombotic agents with poor solubility was developed, which exhibited greatly improved solubility, bioavailability and superior stability, indicating a novel method to overcome the shortages of drug combination.


Subject(s)
Crystallization , Drug Combinations , Pyrazoles/pharmacology , Pyridones/pharmacology , Quercetin/pharmacology , Solvents , Animals , Biological Availability , Calorimetry, Differential Scanning , Chromatography, High Pressure Liquid , Fibrinolytic Agents/pharmacology , Hydrogen Bonding , Male , Pharmaceutical Preparations , Powders , Pyrazoles/chemistry , Pyridones/chemistry , Quercetin/chemistry , Rats , Rats, Sprague-Dawley , Solubility , Temperature , Thermogravimetry , X-Ray Diffraction
7.
Acta Pharmacol Sin ; 42(8): 1223-1234, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33859344

ABSTRACT

Hemorrhagic transformation (HT) is a common serious complication of stroke after thrombolysis treatment, which limits the clinical use of tissue plasminogen activator (t-PA). Since early diagnosis and treatment for HT is important to improve the prognosis of stroke patients, it is urgent to discover the potential biomarkers and therapeutic drugs. Recent evidence shows that pinocembrin, a natural flavonoid compound, exerts anti-cerebral ischemia effect and expands the time window of t-PA. In this study, we investigated the effect of pinocembrin on t-PA-induced HT and the potential biomarkers for HT after t-PA thrombolysis, thereby improving the prognosis of stroke. Electrocoagulation-induced thrombotic focal ischemic rats received intravenous infusion of t-PA (10 mg/kg) 6 h after ischemia. Administration of pinocembrin (10 mg/kg, iv) prior t-PA infusion significantly decreased the infarct volume, ameliorated t-PA-induced HT, and protected blood-brain barrier. Metabolomics analysis revealed that 5 differential metabolites in the cerebral cortex and 16 differential metabolites in serum involved in amino acid metabolism and energy metabolism were significantly changed after t-PA thrombolysis, whereas pinocembrin administration exerted significant intervention effects on these metabolites. Linear regression analysis showed that lactic acid was highly correlated to the occurrence of HT. Further experiments confirmed that t-PA treatment significantly increased the content of lactic acid and the activity of lactate dehydrogenase in the cerebral cortex and serum, and the expression of monocarboxylate transporter 1 (MCT 1) in the cerebral cortex; pinocembrin reversed these changes, which was consistent with the result of metabolomics. These results demonstrate that pinocembrin attenuates HT after t-PA thrombolysis, which may be associated with the regulation of endogenous metabolites. Lactic acid may be a potential biomarker for HT prediction and treatment.


Subject(s)
Cerebral Hemorrhage/drug therapy , Embolic Stroke/drug therapy , Flavanones/therapeutic use , Neuroprotective Agents/therapeutic use , Tissue Plasminogen Activator/therapeutic use , Animals , Biomarkers/blood , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Brain/pathology , Cerebral Hemorrhage/blood , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/pathology , Embolic Stroke/blood , Embolic Stroke/complications , Embolic Stroke/pathology , Lactic Acid/blood , Male , Rats, Sprague-Dawley
9.
Biomed Pharmacother ; 133: 110977, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33249280

ABSTRACT

Puerarin is an isoflavonoid extracted from Pueraria lobate with extensive pharmacological effects in traditional Chinese medicine. The evidence implicates that puerarin mitigates hyperglycemia and various relevant complications. Here, the effect of puerarin on skeletal muscle wasting induced by type 1 diabetes (T1D) was explored. Streptozotocin (STZ)-induced T1D male Sprague Dawley (SD) rats were used in this study. Muscle strength, weight and size were measured. L6 rat skeletal muscle cells were applied for in vitro study. Our results showed that eight-week oral puerarin administration (100 mg/kg) increased muscle strengths and weights accompanied by enhanced skeletal muscle cross-sectional areas in diabetic rats. Simultaneously, puerarin also reduced expressions of several muscle wasting marker genes including F-box only protein 32 (Atrogin-1) and muscle-specific RING-finger 1 (Murf-1) in diabetic group both in vitro and in vivo. Transformation from type I fibers (slow muscle) to type II fibers (fast muscle) were also observed under puerarin administration in diabetic rats. Puerarin promoted Akt/mTOR while inhibited LC3/p62 signaling pathway in skeletal muscle cells. In conclusion, our study showed that puerarin mitigated skeletal muscle wasting in T1D rats and closely related with Akt/mTOR activation and autophagy inhibition. Whether this effect in murine applies to humans remains to be determined.


Subject(s)
Cell Differentiation/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Isoflavones/pharmacology , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Slow-Twitch/drug effects , Muscular Atrophy/prevention & control , Animals , Blood Glucose/metabolism , Cell Line , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/chemically induced , Male , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/pathology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle Strength/drug effects , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Streptozocin , TOR Serine-Threonine Kinases/metabolism , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
10.
Acta Pharmacol Sin ; 42(3): 370-381, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33303991

ABSTRACT

Stroke is an acute cerebrovascular disease caused by ruptured or blocked blood vessels. For the prevention of ischemic stroke, the coagulation state of blood and cerebrovascular protection should be considered. Our previous study has shown that salvianolic acid A (SAA), which is a water-soluble component from the root of Salvia Miltiorrhiza Bge, prevents thrombosis with a mild inhibitory effect on platelet aggregation. In this study we investigated the preventive effects of SAA on cerebrovascular endothelial injury caused by ischemia in vivo and oxygen-glucose deprivation (OGD) in vitro, and explored the underlying mechanisms. An autologous thrombus stroke model was established in SD rats by electrocoagulation. SAA (10 mg/kg) was orally administered twice a day for 5 days before the operation. The rats were sacrificed at 24 h after the operation. We showed that pretreatment with SAA significantly improved the neurological deficits, intracerebral hemorrhage, BBB disruption, and vascular endothelial dysfunction as compared with model group. In human brain microvascular endothelial cells (HBMECs), pretreatment with SAA (10 µM) significantly inhibited OGD-induced cell viability reduction and degradation of tight junction proteins (ZO-1, occludin, claudin-5). Furthermore, we found that SAA inhibited the upregulation of Src signaling pathway in vivo and vitro and reversed the increased expression of matrix metalloproteinases (MMPs) after ischemic stroke. In conclusion, our results suggest that SAA protects cerebrovascular endothelial cells against ischemia and OGD injury via suppressing Src signaling pathway. These findings show that pretreatment with SAA is a potential therapeutic strategy for the prevention of ischemic stroke.


Subject(s)
Caffeic Acids/therapeutic use , Endothelium, Vascular/drug effects , Ischemic Stroke/prevention & control , Lactates/therapeutic use , Neuroprotective Agents/therapeutic use , Signal Transduction/drug effects , Animals , Blood-Brain Barrier/drug effects , Brain/blood supply , Brain/drug effects , Cerebral Hemorrhage/prevention & control , Enzyme Activation/drug effects , Humans , Male , Rats, Sprague-Dawley , Tight Junctions/drug effects , src-Family Kinases/antagonists & inhibitors
11.
Nat Prod Bioprospect ; 10(4): 187-200, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32613339

ABSTRACT

Mangiferin is a compound with many pharmacological activities and exists in many natural products. Anhydrous and hydrate of mangiferin have been reported separately in two literatures, but the polymorphism of this compound has not been realized until this paper. In this study, polymorph screening of mangiferin has been carried out and five forms have been obtained including three new forms never reported. Several solid state characterization methods, such as powder X-ray diffraction, differential scanning calorimetry and thermogravimetry, are used to identify and characterize all of mangiferin forms. The comparison of the crystallographic data and hirshfeld surface analysis were first reported for mangiferin anhydrous and hydrate. Furthermore, the studies on stability, transformation and solubility have been undertaken, the results prompt that form V can be used as the dominant polymorph for the development of innovative pharmaceuticals.

12.
Acta Pharmacol Sin ; 41(4): 516-522, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32047262

ABSTRACT

Excessive nitric oxide (NO) causes extensive damage to the nervous system, and the adrenergic system is disordered in many neuropsychiatric diseases. However, the role of the adrenergic system in protection of the nervous system against sodium nitroprusside (SNP) injury remains unclear. In this study, we investigated the effect of ganoderic acid A (GA A) against SNP injury in neural cells and the role of adrenergic receptors in GA A neuroprotection. We found that SNP (0.125-2 mM) dose-dependently decreased the viability of both SH-SY5Y and PC12 cells and markedly increased NO contents. Pretreatment with GA A (10 µM) significantly attenuated SNP-induced cytotoxicity and NO increase in SH-SY5Y cells, but not in PC12 cells. Furthermore, pretreatment with GA A caused significantly higher adrenaline content in SH-SY5Y cells than in PC12 cells. In order to elucidate the mechanism of GA A-protecting SH-SY5Y cells, we added adrenaline, phentolamine, metoprolol, or ICI 118551 1 h before GA A was added to the culture medium. We found that addition of adrenaline (10 µM) significantly improved GA A protection in PC12 cells. The addition of ß1-adrenergic receptor antagonist metoprolol (10 µM) or ß2-adrenergic receptor antagonist ICI 118551 (0.1 µM) blocked the protective effect of GA A, whereas the addition of α-adrenergic receptor antagonist phentolamine (0.1 µM) did not affect GA A protection in SH-SY5Y cells. These results suggest that ß-adrenergic receptors play an important role in the protection of GA A in SH-SY5Y cells against SNP injuries, and excessive adrenaline system activation caused great damage to the nervous system.


Subject(s)
Heptanoic Acids/pharmacology , Lanosterol/analogs & derivatives , Neurons/drug effects , Neuroprotective Agents/pharmacology , Nitric Oxide/antagonists & inhibitors , Receptors, Adrenergic, beta/metabolism , Animals , Cell Survival/drug effects , Dose-Response Relationship, Drug , Heptanoic Acids/chemistry , Humans , Lanosterol/chemistry , Lanosterol/pharmacology , Molecular Conformation , Neuroprotective Agents/chemistry , Nitric Oxide/metabolism , Nitroprusside/pharmacology , Oxidative Stress/drug effects , PC12 Cells , Rats , Structure-Activity Relationship , Tumor Cells, Cultured
13.
RSC Adv ; 10(55): 33544-33548, 2020 Sep 07.
Article in English | MEDLINE | ID: mdl-35515068

ABSTRACT

Atomic-thin MoS2 materials have attracted increasing attention due to their potentials in numerous fields. However, in 2D-MoS2 sheets, the edge region usually has unique features differing from the interior region, which has potential application in enhancing catalysts and shape-dependent 2D-nanodevices. However, fabricating it cost-effectively is still very difficult. Here, we present one universal method to obtain various shape-dependent closed-edge 2D-MoS2 nanobelts only using one simple step, and width of the MoS2 nanobelts (minimum of 270 nm) were adjustable. Our strategy opens a new fabrication route for closed-edge 2D-MoS2 nanobelts, and in principle, this method is also suitable for other CVD-grown 2D materials.

14.
Acta Pharm Sin B ; 9(3): 505-515, 2019 May.
Article in English | MEDLINE | ID: mdl-31193821

ABSTRACT

Salvianolic acid A (SalA) is an effective compound extracted from traditional Chinese medicine Salvia miltiorrhiza Bunge. The Forkhead box O3a (FOXO3a) signaling pathway plays crucial roles in the modulation of ischemia-induced cell apoptosis. However, no information about the regulatory effect of SalA on FoxO3a is available. To explore the anti-cerebral ischemia effect and clarify the therapeutic mechanism of SalA, SH-SY5Y cells and Sprague-Dawley rats were applied, which were exposed to oxygen glucose deprivation/reoxygenation (OGD/R) and middle cerebral artery occlusion/reperfusion (MCAO/R) injuries, respectively. The involved pathway was identified using the specific inhibitor LY294002. Results showed that SalA concentration-dependently inhibited OGD/R injury triggered cell viability loss. SalA reduced cerebral infarction, lowered brain edema, improved neurological function, and inhibited neuron apoptosis in MCAO/R rats, which were attenuated by the treatment of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) specific inhibitor LY294002. SalA time- and concentration-dependently upregulated the phosphorylation levels of protein kinase B (AKT) and its downstream protein FOXO3a. Moreover, the nuclear translocation of FOXO3a was inhibited by SalA both in vivo and in vitro, which was also reversed by LY294002. The above results indicated that SalA fought against ischemia/reperfusion damage at least partially via the AKT/FOXO3a/BIM pathway.

15.
Int Immunopharmacol ; 63: 299-310, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30142530

ABSTRACT

Neurodegenerative diseases are closely related to neuroinflammation. Drugs targeting inflammation have been proved to be effective in many animal models. Salvianolic acid C (SalC) is a compound isolated from Salvia miltiorrhiza Bunge, a plant with reported effects of inhibiting inflammation. However, the anti-inflammation effects and biological mechanisms of SalC on LPS-stimulated neuroinflammation remain unknown. The aim of this paper was to study its protective effects and its anti-inflammation mechanisms. LPS was used both in vivo and in vitro to induce neuroinflammation in SD rats and microglia cells. MTT assay was carried out to detect cell viability. The levels of TNF­α, IL­1ß, IL­6, IL­10 and PGE2 were detected by ELISA method. The expressions of p­AMPK, p­NF­κB p65, p­IκBα, Nrf2, HO­1 and NQO1 proteins were examined by Western blot analysis. The nuclear translocation of NF­κB p65 was studied by immunofluorescence assay. The specific Nrf2 siRNA was used to clarify the interaction between Nrf2 and NF­κB p65. The AMPK inhibitor Compound C was used study the upstream protein of Nrf2. Results showed that LPS induced the overexpression of inflammatory cytokines and mediated the phosphorylation and nuclear translocation of NF­κB p65 in rat brains and microglia cells. SalC reversed the inflammatory response induced by LPS and inhibited the NF­κB activation. SalC also upregulated the expression of p­AMPK, Nrf2, HO­1 and NQO1. But the anti-inflammation and NF­κB inhibition effects of SalC were attenuated by transfection with specific Nrf2 siRNA or interference with the potent AMPK inhibitor Compound C. In conclusion, SalC inhibited LPS-induced inflammatory response and NF­κB activation through the activation of AMPK/Nrf2 signaling both in vivo and in vitro.


Subject(s)
Alkenes/pharmacology , Anti-Inflammatory Agents/pharmacology , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Polyphenols/pharmacology , AMP-Activated Protein Kinases/metabolism , Alkenes/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , Brain/drug effects , Brain/metabolism , Cell Line , Cytokines/genetics , Cytokines/metabolism , Lipopolysaccharides/pharmacology , Microglia/drug effects , Microglia/metabolism , Polyphenols/therapeutic use , Rats, Sprague-Dawley , Signal Transduction/drug effects
16.
Sensors (Basel) ; 17(1)2017 Jan 23.
Article in English | MEDLINE | ID: mdl-28125000

ABSTRACT

Bistatic forward-looking SAR (BFSAR) is a kind of bistatic synthetic aperture radar (SAR) system that can image forward-looking terrain in the flight direction of an aircraft. Until now, BFSAR imaging theories and methods for a stationary scene have been researched thoroughly. However, for moving-target imaging with BFSAR, the non-cooperative movement of the moving target induces some new issues: (I) large and unknown range cell migration (RCM) (including range walk and high-order RCM); (II) the spatial-variances of the Doppler parameters (including the Doppler centroid and high-order Doppler) are not only unknown, but also nonlinear for different point-scatterers. In this paper, we put forward an adaptive moving-target imaging method for BFSAR. First, the large and unknown range walk is corrected by applying keystone transform over the whole received echo, and then, the relationships among the unknown high-order RCM, the nonlinear spatial-variances of the Doppler parameters, and the speed of the mover, are established. After that, using an optimization nonlinear chirp scaling (NLCS) technique, not only can the unknown high-order RCM be accurately corrected, but also the nonlinear spatial-variances of the Doppler parameters can be balanced. At last, a high-order polynomial filter is applied to compress the whole azimuth data of the moving target. Numerical simulations verify the effectiveness of the proposed method.

17.
Int Immunopharmacol ; 40: 219-228, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27620503

ABSTRACT

Acute lung injury (ALI), characterized by pulmonary edema and inflammatory cell infiltration, is a common syndrome of acute hypoxemic respiratory failure. Methyl salicylate 2-O-ß-d-lactoside (MSL), a natural derivative of salicylate extracted from Gaultheria yunnanensis (Franch.) Rehder, was reported to have potent anti-inflammatory effects on the progression of collagen or adjuvant-induced arthritis in vivo and in vitro. The aim of this study is to investigate the therapeutic effect of MSL on lipopolysaccharide (LPS)-induced acute lung injury and reveal underlying molecular mechanisms. Our results showed that MSL significantly ameliorated pulmonary edema and histological severities, and inhibited IL-6 and IL-1ß production in LPS-induced ALI mice. MSL also reduced MPO activity in lung tissues and the number of inflammatory cells in BALF. Moreover, we found that MSL significantly inhibited LPS-induced TAK1 and NF-κB p65 phosphorylation, as well as the expression of NLRP3 protein in lung tissues. Furthermore, MSL significantly inhibited LPS-induced TAK1 and NF-κB p65 phosphorylation in Raw264.7 cells. In addition, MSL significantly inhibited nuclear translocation of NF-κB p65 in cells treated with LPS in vitro. Taken together, our results suggested that MSL exhibited a therapeutic effect on LPS-induced ALI by inhibiting TAK1/NF-κB phosphorylation and NLRP3 expression.


Subject(s)
Acute Lung Injury/drug therapy , Anti-Inflammatory Agents , Lactose/analogs & derivatives , MAP Kinase Kinase Kinases/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Salicylates , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cell Count , Cytokines/blood , Cytokines/metabolism , Lactose/pharmacology , Lactose/therapeutic use , Lipopolysaccharides , Lung/drug effects , Lung/metabolism , Lung/pathology , MAP Kinase Kinase Kinases/metabolism , Male , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Peroxidase/metabolism , Phosphorylation/drug effects , RAW 264.7 Cells , Salicylates/pharmacology , Salicylates/therapeutic use
18.
Sensors (Basel) ; 16(8)2016 Jul 27.
Article in English | MEDLINE | ID: mdl-27472341

ABSTRACT

Free of the constraints of orbit mechanisms, weather conditions and minimum antenna area, synthetic aperture radar (SAR) equipped on near-space platform is more suitable for sustained large-scene imaging compared with the spaceborne and airborne counterparts. Terrain observation by progressive scans (TOPS), which is a novel wide-swath imaging mode and allows the beam of SAR to scan along the azimuth, can reduce the time of echo acquisition for large scene. Thus, near-space TOPS-mode SAR (NS-TOPSAR) provides a new opportunity for sustained large-scene imaging. An efficient full-aperture imaging scheme for NS-TOPSAR is proposed in this paper. In this scheme, firstly, two-step processing (TSP) is adopted to eliminate the Doppler aliasing of the echo. Then, the data is focused in two-dimensional frequency domain (FD) based on Stolt interpolation. Finally, a modified TSP (MTSP) is performed to remove the azimuth aliasing. Simulations are presented to demonstrate the validity of the proposed imaging scheme for near-space large-scene imaging application.

19.
Cell Physiol Biochem ; 36(1): 395-408, 2015.
Article in English | MEDLINE | ID: mdl-25967977

ABSTRACT

BACKGROUND/AIMS: Diabetes mellitus (DM) characterized by hyperglycemia contributes to macrovascular and microvascular complications. Salvianolic acid A (SalA) is a polyphenolic compound isolated from the root of Salvia miltiorrhiza Bunge, which is a traditional Chinese medicine widely used to treat cardiovascular diseases. However, little is known about its antidiabetic effect. Our study aimed to investigate the in vivo and in vitro antidiabetic effect of SalA and the underlying mechanisms. METHODS: Alloxan-induced type 1 diabetic mice and high-fat diet (HFD) and low-dose streptozotocin (STZ)-induced type 2 diabetic rats received SalA treatment. Blood glucose, oral glucose tolerance test (OGTT), 24-h food and water intake were monitored. In vitro, glucose consumption and uptake were measured in HepG2 cells and L6 myotubes. Mitochondrial function was detected in hepatic and skeletal muscle mitochondria. AMP-activated protein kinase (AMPK) and Akt were analyzed by western blot. RESULTS: In both type 1 and type 2 diabetic animals, SalA lowered fasting blood glucose (FBG) and fed blood glucose in dose-dependent manner, as well as reduced 24-h food and water intake. In vitro, SalA caused dose-dependent increase in glucose consumption and enhanced glucose uptake. SalA significantly increased ATP production from 10 min to 12 h in HepG2 cells and L6 myotubes. Interestingly, SalA decreased mitochondrial membrane potential (MMP) in HepG2 cells. Furthermore, SalA improved hepatic and skeletal muscle mitochondrial function, increased ATP production, and concurrently decreased MMP. In particularly, SalA activated AMPK phosphorylation through Ca(2+)/calmodulin-dependent protein kinase kinase ß (CaMKKß)/AMPK signaling pathway, independent of liver kinase 1 (LKB1)/AMPK pathway. However, SalA didn't show any effect on insulin secretagogue and activation of PI3K/Akt signaling pathway. CONCLUSION: SalA exhibits the antidiabetic effects in diabetic animal models through improving mitochondrial function, increasing ATP production, and decreasing MMP via CaMKKß/AMPK signaling pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Alkenes/administration & dosage , Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/administration & dosage , Mitochondria/drug effects , Polyphenols/administration & dosage , Alkenes/pharmacology , Alloxan , Animals , Blood Glucose/drug effects , Cell Line , Diabetes Mellitus, Experimental/metabolism , Diet, High-Fat , Eating/drug effects , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Hypoglycemic Agents/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/metabolism , Polyphenols/pharmacology , Rats , Signal Transduction/drug effects , Streptozocin
20.
Am J Chin Med ; 42(5): 1183-98, 2014.
Article in English | MEDLINE | ID: mdl-25183303

ABSTRACT

Type 2 diabetes patients have an increased risk of developing hepatic fibrosis. Salvianolic acid A (SalA) has been reported to be a strong polyphenolic anti-oxidant and free radical scavenger. The aim of the present study was to evaluate the effect of SalA on the pathological progression of hepatic fibrosis in high-fat diet (HFD)-fed and streptozotocin (STZ)-induced diabetic rats and to clarify the underlying mechanisms. Type 2 diabetic animal model with hepatic fibrosis was developed by a high-sucrose, HFD and low-dose STZ injection (i.p.). Diabetic rats were randomly divided into SalA group (0.3 mg/kg/day) and diabetic control groups fed with a HFD. After administration for four months, SalA reversed the hyperlipidemia and reduced hepatic triglyceride (TG). Hematoxylin-Eosin (HE) and Picro acid-Sirius red staining results indicated that SalA significantly alleviated the lesions of hepatic steatosis and fibrosis, with the reduction of type I and III collagens. The expression of α-smooth-muscle-actin (α-SMA) and transforming growth factor ß1 (TGF-ß1) in the liver were markedly down-regulated by SalA treatment. TUNEL staining showed that SalA reduced apoptosis in hepatocytes. In addition, SalA improved hepatic mitochondrial respiratory function in diabetic rats. Taken together, these findings demonstrated that SalA could prevent the pathological progression of hepatic fibrosis in HFD-fed and STZ-induced diabetic rats. The underlying mechanisms may be involved in reducing oxidative stress, suppressing α-SMA and TGF-ß1 expression, as well as exerting anti-apoptotic and mitochondria-protective effects.


Subject(s)
Alkenes/therapeutic use , Caffeic Acids/therapeutic use , Diabetes Mellitus, Experimental/complications , Diet, High-Fat/adverse effects , Lactates/therapeutic use , Liver Cirrhosis/etiology , Liver Cirrhosis/prevention & control , Phytotherapy , Polyphenols/therapeutic use , Actins/metabolism , Alkenes/administration & dosage , Animals , Caffeic Acids/administration & dosage , Disease Progression , Lactates/administration & dosage , Liver/metabolism , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Polyphenols/administration & dosage , Rats, Sprague-Dawley , Streptozocin , Transforming Growth Factor beta1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL