Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Phytother Res ; 37(7): 2787-2799, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36807664

ABSTRACT

Pulmonary fibrosis (PF) is a progressive and fatal interstitial lung disease with limited therapeutic options at present, and epithelial-mesenchymal transition (EMT) is recognized as a major cause of lung fibrosis. Our previous work has confirmed that total extract of Anemarrhena asphodeloides Bunge [Asparagaceae] exerted the effect of anti-PF. As a main constituent of Anemarrhena asphodeloides Bunge [Asparagaceae], the effect of timosaponin BII (TS BII) on drug-induced EMT process in PF animals and alveolar epithelial cells remains unknown. In this study, we evaluated the effect of TS BII on bleomycin (BLM)-induced PF. The results showed that TS BII could restore the structure of lung architecture and MMP-9/TIMP-1 balance in fibrotic rat lung and inhibit collagen deposition. Moreover, we found that TS BII could reverse the abnormal expression of TGF-ß1 and EMT-related marker proteins including E-cadherin, vimentin, and α-SMA. Besides, aberrant TGF-ß1 expression and phosphorylation of Smad2 and Smad3 in BLM-induced animal model and TGF-ß1-induced cell model were downregulated by TS BII treatment, indicating that EMT in fibrosis was suppressed by inhibition of TGF-ß/Smad pathway both in vivo and in vitro. In summary, our study suggested that TS BII could be a promising candidate for PF treatment.


Subject(s)
Pulmonary Fibrosis , Rats , Animals , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1/metabolism , Epithelial-Mesenchymal Transition , Lung , Fibrosis , Bleomycin/adverse effects
2.
Eur J Med Chem ; 245(Pt 1): 114860, 2023 Jan 05.
Article in English | MEDLINE | ID: mdl-36370550

ABSTRACT

In order to take advantage of both immunotherapeutic and metabolic antitumor agents, novel dual indoleamine 2,3- dioxygenase 1 (IDO1) and thioredoxin reductase 1 (TrxR1) inhibitors were designed. Thioredoxin reductase 1 (TrxR1) is a main ROS modulator within CRC cells. Indoleamine 2,3-dioxygenase (IDO1) is crucial controller for tryptophan (Trp) metabolism that is also important for CRC immunotherapy. Herein, ten compounds 12a-j containing hydroxyamidine scaffold were designed, synthesized and evaluated for inhibitory activities against IDO1/TrxR1 enzyme and CRC cells. Among these compounds, the most active compound 12d (ZC0109) showed excellent and balanced activity against both IDO1 (IC50 = 0.05 µM) and TrxR1 (IC50 = 3.00 ± 0.25 µM) were selected for further evaluation. Compound ZC0109 exhibited good dual inhibition against IDO1 and TrxR1 both in vitro and in vivo. Further mechanistic studies reveal that, through IDO1 and TrxR1 inhibition by ZC0109 treatment, accumulated ROS effectively induced apoptosis and G1/S cell cycle arrest in cancer cells. In vivo evaluation demonstrated excellent anti-tumor effect of ZC0109 with the notable ability of promoting ROS-induced apoptosis, reducing kynurenine level in plasma and restoring anti-tumor immune response. Thus, ZC0109 represents a potential CRC therapy agent for further development.


Subject(s)
Colorectal Neoplasms , Enzyme Inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase , Reactive Oxygen Species , Thioredoxin Reductase 1 , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Thioredoxin Reductase 1/antagonists & inhibitors , Cell Line, Tumor , Humans , Apoptosis/drug effects , Colorectal Neoplasms/enzymology
3.
J Anal Methods Chem ; 2022: 6209731, 2022.
Article in English | MEDLINE | ID: mdl-35912019

ABSTRACT

A simple, sensitive, and fluorescent immunoassay for the detection of prostate-specific antigen (PSA) based on horseradish peroxidase and silicon dioxide nanospheres as a signal amplification strategy has been described. In the design, the primary antibody (Ab1) of PSA was first immobilized on the 96-well plates via physical adsorption between polystyrene and hydrophobic groups of the antibody molecule. The silicon dioxide nanospheres (SiO2 NSs), with large surface area and good biocompatibility, were loaded with horseradish peroxidase (HRP) and horseradish peroxidase-labeled secondary antibodies (HRP-Ab2) as signal amplification systems. In the presence of PSA, a sandwich-type immunocomplex composed of Ab1-Ag-HRP-Ab2 had been formed. Fluorescence technology was employed to obtain the response signal of the immunoassay in the L-tyrosine and H2O2 systems. Experiment results showed that a strong and stable fluorescent signal at 416 nm (excitation wavelength: 325 nm) was observed, and the changes in fluorescent intensity were related to the levels of PSA. Under the optimal conditions, the relative fluorescence intensity was linear with the logarithm of PSA concentration from 0.03 to 100 ng·mL-1, with a detection limit of 0.01 ng·mL-1 (at a signal/noise ratio of 3). In contrast to other fluorescent immunoassays, the sandwich-type immunoreaction based on the high binding ELISA plates has the advantages of being simple, stable, and easy to operate, high selectivity, small sample quantity, etc., which can be widely used in the selective detection of a variety of targets, from DNA to proteins and small molecules. Such fluorescent immunoassays should be feasible for the fields of molecular diagnosis and other life science fields in the future.

4.
BMC Anesthesiol ; 22(1): 225, 2022 07 16.
Article in English | MEDLINE | ID: mdl-35842610

ABSTRACT

OBJECTIVE: To investigate the analgesic effect and rehabilitation quality of patients undergoing thoracoscopic wedge resection of the lung under erector spinae plane (ESP) block with dexmedetomidine combined with the same dose and different concentrations of ropivacaine. METHODS: Seventy patients undergoing thoracoscopic wedge resection were randomly divided into groups A (n = 35) and B (n = 35). To perform ESP block, the groups were administered dexmedetomidine (0.5 µg/kg) combined with 30 mL of 0.33% ropivacaine or 20 mL of 0.5% ropivacaine, respectively, half an hour before general anesthesia induction. We compared the onset time of anesthesia, the block level, and the duration of the block between the two groups. The number of compressions of the analgesic pump within 24 h and 48 h postoperatively and the time of the first compression were noted. The visual analog scale (VAS) scores of static and cough at 0.5 h, 6 h, 12 h, 24 h, and 48 h postoperatively were noted. Furthermore, the 40-item quality of recovery questionnaire (QoR-40) score was recorded at 24 h postoperatively. In addition, we noted the time taken to get out of the bed for the first time, the length of hospital stay, analgesia satisfaction, and the occurrence of related adverse reactions and complications within 48 h postoperatively. RESULTS: The range of ESP block was wider in Group A than in Group B (P < 0.05). Group B had a significantly shorter onset time (P < 0.05) and lower static and cough VAS scores at 6 h and 12 h postoperatively (P < 0.05); this was associated with significantly fewer compressions of the analgesic pump within 24 h and 48 h postoperatively and significantly more time until the first compression of the analgesic pump was required (P < 0.05). Group B was associated with significantly superior QoR-40 scores 24 h postoperatively and significantly shorter time to get out of the bed for the first time than Group A (P < 0.05). CONCLUSION: Dexmedetomidine combined with 0.5% ropivacaine for ESP block is better than 0.33% ropivacaine for overall analgesia and postoperative rehabilitation of patients undergoing thoracoscopic wedge resection. TRIAL REGISTRATION: ChiCTR2200058114 , Date of registration: 30/03/2022.


Subject(s)
Dexmedetomidine , Nerve Block , Anesthetics, Local , Cough/etiology , Humans , Lung , Nerve Block/adverse effects , Pain, Postoperative/etiology , Pain, Postoperative/prevention & control , Prospective Studies , Ropivacaine , Ultrasonography, Interventional
5.
Bioorg Chem ; 119: 105546, 2022 02.
Article in English | MEDLINE | ID: mdl-34954573

ABSTRACT

Pulmonary fibrosis is a progressive interstitial lung disease with poor prognosis. Anemarrhenae Rhizoma is a traditional Chinese herbal medicine and has been applied in clinical practice for a long history. Recently, components of Anemarrhenae Rhizoma were reported to possess anti-inflammatory and immunomodulatory features; however, the effect of them on pulmonary fibrosis remains unknown. In this study, we explored the therapeutic effect of total extract of Anemarrhenae Rhizoma (TEAR) on bleomycin-induced pulmonary fibrosis. Pulmonary fibrosis rat model was established by a single intratracheal instillation of bleomycin, three doses of TEAR were intragastrically administered for consecutive 28 days. Subsequent to sacrificing of rats, pulmonary fibrosis was observed in rats treated with bleomycin, but administration of TEAR attenuated lung fibrosis, as evidenced by the improved lung histopathological damage and decreased weight loss and lung index. Moreover, TEAR treatment inhibited the inflammatory response in lung fibrosis, which was shown by the reduced nitrogen oxide level and myeloperoxidase activity. Furthermore, TEAR modulated the redox balance in lung tissue by alleviated lipid peroxidation and enhanced enzymatic antioxidants activity. Meanwhile, TEAR protected the rats from fibrosis in a dose-dependent manner, and the anti-fibrotic activity of TEAR may be related to the modulation of TGF-ß1/Smad signaling pathway. Collectively, TEAR alleviates bleomycin-induced pulmonary fibrosis, indicating perspectives for development of a potential agent for lung fibrosis therapy.


Subject(s)
Anemarrhena/chemistry , Drugs, Chinese Herbal/therapeutic use , Flavonoids/therapeutic use , Pulmonary Fibrosis/drug therapy , Rhizome/chemistry , Saponins/therapeutic use , Animals , Bleomycin , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/isolation & purification , Flavonoids/chemistry , Flavonoids/isolation & purification , Male , Medicine, Chinese Traditional , Molecular Structure , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Rats , Rats, Sprague-Dawley , Saponins/chemistry , Saponins/isolation & purification , Structure-Activity Relationship
6.
Drug Deliv ; 26(1): 499-508, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31033359

ABSTRACT

In this paper, the self-assembled folate-biotin-quaternized starch nanoparticles (FBqS NPs) were used as carrier system of doxorubicin (DOX) and siRNAIGF1R for the codelivery of both into human lung adenocarcinoma cell lines (A549 cells) in vitro. The cytotoxicity, targeted ligand competition, cell proliferation inhibition, cellular uptake, endocytosis mechanism and target protein suppression of drug-loaded FBqS NPs were evaluated in detail. Compared with several other drug formulations under same condition, siRNAIGF1R/DOX/FBqS NPs exhibited the greatest cytotoxicity to A549 cells and the cytotoxicity was competitively inhibited by free folate in dose-dependent manner. The A549 cells treated by siRNAIGF1R/DOX/FBqS NPs showed the lowest cell proliferation capacity. The energy-dependent clathrin- and caveolae-mediated endocytosis might be the primary cellular uptake mechanism of drug-loaded FBqS NPs. The expression of IGF1R protein in A549 cells treated by siRNAIGF1R/FBqS NPs declined dramatically. So the FBqS NPs were expected as the co-carrier system of chemotherapeutants and siRNAs for future clinical application.


Subject(s)
Antineoplastic Agents/administration & dosage , Doxorubicin/administration & dosage , Drug Carriers/chemistry , Nanoparticles/chemistry , RNA, Small Interfering/administration & dosage , A549 Cells , Antineoplastic Agents/pharmacology , Biotin/chemistry , Cell Survival/drug effects , Doxorubicin/pharmacology , Drug Synergism , Endocytosis/drug effects , Folic Acid/chemistry , Humans , Lung Neoplasms/pathology , Particle Size , RNA, Small Interfering/pharmacology , Starch/chemistry
7.
Artif Cells Nanomed Biotechnol ; 46(sup3): S774-S782, 2018.
Article in English | MEDLINE | ID: mdl-30280608

ABSTRACT

In this paper, a new oral insulin formulation, insulin-loaded carboxymethyl-ß-cyclodextrin-grafted chitosan nanoparticles (insulin/CMCD-g-CS NPs), was fabricated by ionic crosslinking technique. The therapeutic efficacy of new formulation was investigated in detail. Firstly, the CMCD-g-CS was synthesized by EDC-mediated esterification reaction. The prepared CMCD-g-CS exhibited favourable loading capacity and encapsulation efficiency of drug. The release experiment in vitro showed that the nanocarrier could efficiently protect encapsulated insulin at simulated gastric environment and release drug in the simulated colonic fluid. The insulin/CMCD-g-CS NPs effectively promoted drug internalization into Caco-2 cells and could reversibly open the tight junction between cells. The oral administration of insulin/CMCD-g-CS NPs could lastingly decrease blood sugar level in diabetic mice. The liver function study verified that the insulin/CMCD-g-CS NPs had not obvious toxicity to experimental mice. Therefore, the CMCD-g-CS could be an effective and safe oral insulin delivery carrier for future clinical application. A new biocompatible polysaccharide nanoparticle was fabricated as oral insulin delivery carrier for improving diabetic treatment.


Subject(s)
Chitosan , Diabetes Mellitus, Experimental , Drug Carriers , Insulin , beta-Cyclodextrins , Administration, Oral , Animals , Caco-2 Cells , Chitosan/chemistry , Chitosan/pharmacokinetics , Chitosan/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Drug Liberation , Humans , Insulin/chemistry , Insulin/pharmacokinetics , Insulin/pharmacology , Male , Mice , beta-Cyclodextrins/chemistry , beta-Cyclodextrins/pharmacokinetics , beta-Cyclodextrins/pharmacology
8.
Reg Anesth Pain Med ; 43(8): 838-843, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29923955

ABSTRACT

BACKGROUND AND OBJECTIVES: Although intravenous lipid emulsion has been proved a powerful antidote for local anesthetic toxicity, there are few pharmacokinetic data on using lipid infusion as a pretreatment for other clinical applications. We assessed the influence of lipid pretreatment on the pharmacodynamics and pharmacokinetics of levobupivacaine. METHODS: Altogether, 12 patients undergoing below-knee surgery for a fracture were randomly assigned to 2 groups (6 patients per group): pretreatment with 1.5 mL/kg lipid infusion (lipid group) or saline infusion (control subjects) followed by complete femoral and sciatic nerve block with 0.375% levobupivacaine (2.5 mg/kg). Total and free (non-protein bound) plasma levobupivacaine concentrations and triglycerides in the lipid group were determined. RESULTS: Results were given as means ± SD. Total and free maximum plasma levobupivacaine concentrations were lower in the lipid group than in control subjects (865 ± 98 vs 1145 ± 177 µg/L and 56.8 ± 7.5 vs 78.2 ± 13.7 µg/L, respectively; P < 0.01). Apparent volume of distribution and clearance were higher in the lipid group than in control subjects (211 ± 35 vs 170 ± 21 L and 35.1 ± 8.0 vs 25.8 ± 2.6 L/h, respectively; P < 0.05). Triglyceride level was significantly higher at the end of lipid infusion than baseline values (7.59 ± 1.32 vs 1.34 ± 0.39 mmol/L; P < 0.01). CONCLUSIONS: Lipid pretreatment increased the apparent volume of distribution and clearance and decreased the maximum total and free levobupivacaine concentrations, thus offering a reasonable explanation for the effects of lipids on local anesthesia-related toxicity in humans. Rapid lipid infusion induced hypertriglyceridemia without other apparent risks in this study. CLINICAL TRIAL REGISTRATION: This study was registered at the Chinese Clinical Trial Registry, identifier ChiCTR-TRC-14005203.


Subject(s)
Anesthetics, Local/blood , Autonomic Nerve Block/methods , Fat Emulsions, Intravenous/administration & dosage , Femoral Nerve/drug effects , Fractures, Bone/blood , Levobupivacaine/blood , Sciatic Nerve/drug effects , Adult , Anesthetics, Local/administration & dosage , Female , Femoral Nerve/physiology , Fractures, Bone/surgery , Humans , Levobupivacaine/administration & dosage , Male , Middle Aged , Pain Measurement/drug effects , Pain Measurement/methods , Pain, Postoperative/blood , Pain, Postoperative/prevention & control , Preoperative Care/methods , Sciatic Nerve/physiology
9.
J Pharm Pharmacol ; 68(7): 941-52, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27139338

ABSTRACT

OBJECTIVES: The aim of this study was to investigate the cardioprotective effect of salvianolic acid B (Sal B) on acute myocardial infarction (AMI) in rats and its potential mechanisms. METHODS: The AMI model was established in rats to study the effect of Sal B on AMI. Haematoxylin-eosin (HE) staining was used to evaluate the pathological change in AMI rats. Immunofluorescence and TUNEL staining were used to detect autophagy and apoptosis of myocardial cells in hearts of AMI rats, respectively. Protein expression of apoptosis-related, autophagy-related and angiogenesis-related proteins were examined by Western blot. KEY FINDINGS: Sal B attenuated myocardial infarction significantly compared with that of the model group. Rats administered with Sal B showed higher inhibition rate of infarction and lower infarct size than those of the model group. Moreover, Sal B decreased the serum levels of creatine kinase, lactate dehydrogenase and malondialdehyde, while increased such level of superoxide dismutase significantly compared with those of the model group. Sal B inhibited the expression of Bax, cleaved caspase-9 and cleaved PARP, while promoted the expression of Bcl-2, LC3-II, Beclin1 and VEGF. CONCLUSIONS: Sal B has cardioprotective effect on AMI and Sal B may be a promising candidate for AMI treatment.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Benzofurans/pharmacology , Cardiotonic Agents/pharmacology , Myocardial Infarction/prevention & control , Neovascularization, Physiologic/drug effects , Animals , Apoptosis Regulatory Proteins/biosynthesis , Beclin-1/biosynthesis , Creatine Kinase/blood , L-Lactate Dehydrogenase/blood , Male , Malondialdehyde/blood , Microtubule-Associated Proteins/biosynthesis , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocardium/pathology , Rats , Superoxide Dismutase/blood , Vascular Endothelial Growth Factor A/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...