Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Mol Pharm ; 17(3): 757-768, 2020 03 02.
Article in English | MEDLINE | ID: mdl-32011888

ABSTRACT

Salmon calcitonin (sCT) is a potent calcium-regulating peptide hormone and widely applied for the treatment of some bone diseases clinically. However, the therapeutic usefulness of sCT is hindered by the frequent injection required, owing to its short plasma half-life and therapeutic need for a high dose. Oral delivery is a popular modality of administration for patients because of its convenience to self-administration and high patient compliance, while orally administered sCT remains a great challenge currently due to the existence of multiple barriers in the gastrointestinal (GI) tract. Here, we introduced an orally targeted delivery system to increase the transport of sCT across the intestine through both the paracellular permeation route and the bile acid pathway. In this system, sCT-based glycol chitosan-taurocholic acid conjugate (GC-T)/dextran sulfate (DS) ternary nanocomplexes (NC-T) were produced by a flash nanocomplexation (FNC) process in a kinetically controlled mode. The optimized NC-T exhibited well-controlled properties with a uniform and sub-60 nm hydrodynamic diameter, high batch-to-batch reproducibility, good physical or chemical stability, as well as sustained drug release behaviors. The studies revealed that NC-T could effectively improve the intestinal uptake and permeability, owing to its surface functionalization with the taurocholic acid ligand. In the rat model, orally administered NC-T showed an obvious hypocalcemia effect and a relative oral bioavailability of 10.9%. An in vivo assay also demonstrated that NC-T induced no observable side effect after long-term oral administration. As a result, the orally targeted nanocomplex might be a promising candidate for improving the oral transport of therapeutic peptides.


Subject(s)
Calcitonin/administration & dosage , Calcium-Regulating Hormones and Agents/administration & dosage , Drug Delivery Systems/methods , Intestinal Absorption/drug effects , Nanocomposites/chemistry , Administration, Oral , Animals , Biological Availability , Biological Transport , Caco-2 Cells/drug effects , Caco-2 Cells/metabolism , Calcitonin/adverse effects , Calcitonin/blood , Calcitonin/pharmacokinetics , Calcium/blood , Calcium-Regulating Hormones and Agents/adverse effects , Calcium-Regulating Hormones and Agents/blood , Calcium-Regulating Hormones and Agents/pharmacokinetics , Chitosan/chemistry , Dextran Sulfate/chemistry , Drug Liberation , Drug Stability , Half-Life , Humans , Hypocalcemia/chemically induced , Injections, Subcutaneous , Male , Rats , Rats, Sprague-Dawley , Taurocholic Acid/chemistry
2.
Cell Death Differ ; 27(6): 1924-1937, 2020 06.
Article in English | MEDLINE | ID: mdl-31827236

ABSTRACT

Acetaminophen (APAP) is the leading cause of drug-induced acute liver failure. Sphingosine-1-phosphate (S1P), whose formation is catalyzed by sphingosine kinase (SPHK)-1 or -2, is a bioactive lipid implicated in human health and disease. Here, we show that APAP-treated sphK1-deficient (sphK1-/-) mice exhibited markedly less liver damage and reduced inflammation compared with the wild-type mice. SPHK1 deficiency alleviated APAP-induced endoplasmic reticulum (ER) stress by affecting the phosphorylation of inositol-requiring enzyme 1α (IRE1α) and protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK)-eukaryotic translation initiation factor 2α (eIF2α), levels of activating transcription factor 4 (ATF4), and activation of activating transcription factor 6 (ATF6). SPHK1 deficiency also inhibited mitochondrial permeability transition (MPT), as evidenced by the impaired phosphorylation of JNK, apoptosis signal-regulated kinase 1 (ASK1), and glycogen synthase kinase 3ß (GSK3ß). In addition, SPHK1 deficiency reduced the levels of histone deacetylase and promoted the acetylation of p65 and STAT1, thereby impairing the transcription of inflammatory genes. Supplementation with exogenous S1P significantly reversed the activation of the PERK-eIF2α-ATF4 pathway and ATF6 during ER stress as well as the activation of GSK3ß, ASK1, and JNK during MPT. Both FTY720, a functional S1P receptor antagonist, and PF543, an SPHK1 inhibitor, significantly ameliorated APAP-induced liver injury and improved animal survival. Our study reveals a critical role for SPHK1 in mediating APAP-induced hepatotoxicity by promoting ER stress and MPT.


Subject(s)
Acetaminophen/toxicity , Endoplasmic Reticulum Stress/drug effects , Fingolimod Hydrochloride/pharmacology , Mitochondrial Transmembrane Permeability-Driven Necrosis/drug effects , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Pyrrolidines/pharmacology , Sulfones/pharmacology , Animals , Enzyme Inhibitors , Methanol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors
3.
Phytomedicine ; 62: 152965, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31129432

ABSTRACT

BACKGROUND: Anti-malarial drug artesunate (ART), a semi-synthetic derivative of artemisnin, has immunosuppressive effects on several autoimmune diseases, including Systemic lupus erythematosus (SLE), Rheumatoid arthritis (RA), and Colitis. However, molecular mechanisms of ART, especially on follicular helper T cells (Tfh), central players in SLE pathology, are far from clear. PURPOSE: The object for this work is to investigate the therapeutic effect of ART on lupus-prone MRL/lpr mice and its regulatory function on Tfh cells. STUDY DESIGN AND METHODS: MRL/lpr mice were used to explore therapeutic effects of ART on lupus-prone MRL/lpr mice and its regulatory functions on Tfh cells. Then, experiments of renal function were accomplished using the biochemical kits. Effects of ART on histopathology of kidneys, inflammatory factors and autoantibodies were examined using H&E staining, ELISA and real-time PCR. Flow cytometry and western blot analysis were used to examine effects of ART on Tfh differentiation and Jak2-Stat3 signaling pathway. RESULTS: Upon oral administration, ART significantly prolonged the survival of MRL/lpr mice, ameliorated the lupus nephritis symptoms, decreased the levels of anti-dsDNA antibodies deposited in the kidney, and the levels of pathogenic cytokines (IL-6, IFN-γ and IL-21). After ART treatment, T-cell compartment in the spleen of MRL/lpr mice was restored in terms of reduction in the number of Tfh cells and in the maintenance of the ratio of Tfr to follicular regulatory T cells (Tfh). In addition, ART has significantly inhibited the phosphorylation levels of Jak2 and Stat3 in the MRL/lpr mice. CONCLUSION: ART showed therapeutic effects on lupus-prone MRL/lpr mice by inhibiting the differentiation of Tfh cells as well as altering the activation status of Jak2-Stat3 signaling cascade.


Subject(s)
Antimalarials/pharmacology , Artesunate/pharmacology , Lupus Erythematosus, Systemic/drug therapy , Lupus Nephritis/drug therapy , Signal Transduction/drug effects , Animals , Autoantibodies/drug effects , Cell Differentiation/drug effects , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Janus Kinase 2/metabolism , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred MRL lpr , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology
4.
J Neuroimmune Pharmacol ; 14(3): 478-492, 2019 09.
Article in English | MEDLINE | ID: mdl-31069623

ABSTRACT

Parkinson's disease (PD) is characterized by the degeneration of dopaminergic neurons and excessive microglial activation in the substantia nigra pars compacta (SNpc). In the present study, we aimed to demonstrate the therapeutic effectiveness of the potent sphingosine-1-phosphate receptor antagonist fingolimod (FTY720) in an animal model of PD induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and to identify the potential mechanisms underlying these therapeutic effects. C57BL/6J mice were orally administered FTY720 before subcutaneous injection of MPTP. Open-field and rotarod tests were performed to determine the therapeutic effect of FTY720. The damage to dopaminergic neurons and the production of monoamine neurotransmitters were assessed using immunohistochemistry, high-performance liquid chromatography, and flow cytometry. Immunofluorescence (CD68- positive) and enzyme-linked immunosorbent assay were used to analyze the activation of microglia, and the levels of activated signaling molecules were measured using Western blotting. Our findings indicated that FTY720 significantly attenuated MPTP-induced behavioral deficits, reduced the loss of dopaminergic neurons, and increased dopamine release. FTY720 directly inhibited MPTP-induced microglial activation in the SNpc, suppressed the production of interleukin (IL)-6, IL-1ß, and tumor necrosis factor-α in BV-2 microglial cells treated with 1-methyl-4-phenylpyridinium (MPP+), and subsequently decreased apoptosis in SH-SY5Y neuroblastoma cells. Moreover, in MPP+-treated BV-2 cells and primary microglia, FTY720 treatment significantly attenuated the increases in the phosphorylation of PI3K/AKT/GSK-3ß, reduced ROS generation and p65 activation, and also inhibited the activation of NLRP3 inflammasome and caspase-1. In conclusion, FTY720 may reduce PD progression by inhibiting NLRP3 inflammasome activation via its effects on ROS generation and p65 activation in microglia. These findings provide novel insights into the mechanisms underlying the therapeutic effects of FTY720, suggesting its potential as a novel therapeutic strategy against PD. Graphical Abstract FTY720 may reduce ROS production by inhibiting the PI3K/AKT/GSK-3ß signaling pathway, while at the same time reducing p65 phosphorylation, thus decreasing NLRP3 inflammasome activation through these two pathways, ultimately reducing microglia activation-induced neuronal damage.


Subject(s)
Antiparkinson Agents/pharmacology , Fingolimod Hydrochloride/pharmacology , Inflammasomes/drug effects , Microglia/drug effects , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/drug therapy , Animals , Apoptosis/drug effects , Cell Line , Corpus Striatum/chemistry , Corpus Striatum/drug effects , Cytokines/biosynthesis , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Drug Evaluation, Preclinical , Exploratory Behavior/drug effects , Inflammasomes/metabolism , MPTP Poisoning/drug therapy , MPTP Poisoning/immunology , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Mitochondria/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Parkinsonian Disorders/immunology , Pars Compacta/chemistry , Pars Compacta/drug effects , Pars Compacta/pathology , Reactive Oxygen Species , Rotarod Performance Test , Signal Transduction/drug effects
5.
J Ethnopharmacol ; 238: 111861, 2019 Jun 28.
Article in English | MEDLINE | ID: mdl-30954617

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Wang-bi tablet (WB) consists of 17 traditional Chinese medicines and has been used for treating rheumatoid arthritis (RA) in China for many years, however, its pharmacologic mechanism is not clear. AIM OF STUDY: The aim of this study was to investigate the therapeutic effect of WB on collagen-induced mouse arthritis and explored the underlying mechanism. MATERIALS AND METHODS: DBA/1 mice were used to establish a type II collagen-induced arthritis (CIA) model. From the day of arthritis onset, mice were treated daily by gavage with either total glucosides of paeony (TGP, 0.37  g/kg/d) or WB at a lower (1.11  g/kg/d, WBL) or higher dose of (3.33  g/kg/d, WBH) for 8 weeks. The severity of arthritis, levels of cytokines and the activation of signaling pathways were determined. RESULTS: Our results revealed that WB treatment effectively alleviated inflammatory symptoms and prevented bone erosions and joint destructions. It obviously decreased the serum concentration of pro-inflammatory cytokines TNF-α, IL-6 and IL-17α, while increased the concentration of anti-inflammatory cytokine IL-10. Interestingly, the proportion of splenic Treg cells were increased significantly. In vitro experiments showed that WB inhibited the differentiation of osteoclasts. Consistently, the mRNA levels of tartrate-resistant acid phosphatase (TRAP) and cathepsin K (CtsK), and the activation of NF-κB and JAK-STAT3 signaling pathways in the paws of CIA mice were inhibited by WB treatment. On the other hand, up-regulation of osteogenic genes Runx2, Osterix mRNA, and activation of Wnt/ß-catenin signaling pathway along with a decreased receptor activator of nuclear factor κB ligand (RANKL) expression were found in WB treated mice. CONCLUSION: Our results suggest that the therapeutic effect of Wang-bi tablet could be attributed to its inhibitory activity on NF-κB and STAT3 signaling pathway-mediated osteoclast differentiation, and its enhancement on Wnt/ß-catenin signaling pathway-mediated osteoblast functions.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/drug therapy , Drugs, Chinese Herbal/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Bone and Bones/drug effects , Bone and Bones/pathology , Cytokines/immunology , Drugs, Chinese Herbal/pharmacology , Joints/drug effects , Joints/pathology , Male , Medicine, Chinese Traditional , Mice, Inbred DBA , NF-kappa B/metabolism , Osteoblasts/drug effects , Osteoblasts/physiology , Osteoclasts/drug effects , Osteoclasts/physiology , RANK Ligand/metabolism , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Wnt Signaling Pathway/drug effects
6.
Int J Mol Med ; 42(3): 1666-1674, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29901091

ABSTRACT

Rheumatoid arthritis (RA) severely affects the quality life of patients due to its high association with disability. Traditional Chinese medicines have been reported to exert notable therapeutic effects on RA. The Chinese medicinal prescription Wang­Bi Tablet (WB) has been successfully used to clinically treat RA for many years; however, its pharmacological mechanism of action is largely unclear. In the present study, adjuvant­induced arthritis (AIA) rats were used to evaluate the anti­inflammatory effects of WB and western blotting was used to explore the molecular mechanisms. The experimental results demonstrated that WB treatment significantly reduced arthritis score and hind­paw volume. Furthermore, synovial hyperplasia, inflammatory cell infiltration and joint destruction were ameliorated by WB. The expression levels of the proinflammatory cytokines interleukin (IL)­1ß, tumor necrosis factor­α and IL­6, were reduced in the joints of WB­treated rats. Western blotting revealed that WB could also inhibit excessive activation of nuclear factor (NF)­κB and Janus kinase (JAK)­signal transducer and activator of transcription 3 (STAT3) signaling pathways. These results indicated that the therapeutic effects of WB on AIA may be accomplished through inhibition of the NF­κB and JAK­STAT3 signaling pathways. These findings provide experimental evidence to support WB as a therapeutic agent for the treatment of patients with RA.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Drugs, Chinese Herbal/therapeutic use , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Animals , Arthritis, Experimental/pathology , Cytokines/metabolism , Drugs, Chinese Herbal/pharmacology , Inflammation Mediators/metabolism , Joints/drug effects , Joints/pathology , Lipopolysaccharides , Male , Mice , Protective Agents/pharmacology , RAW 264.7 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley
7.
Biomaterials ; 178: 620-629, 2018 09.
Article in English | MEDLINE | ID: mdl-29602561

ABSTRACT

Morphology of delivery nanovehicle plays a significant role in bioavailability of drug. Molecular bottlebrush (MBB)-based unimolecular micelle, with tunable morphologies including sphere, rod, and worm, offers a new aspect to uncover the relationship between morphology and bio-behaviors. In this study, a series of MBB as unimolecular micelle with core-shell structures were tailor-made through controlled/living polymerization and click chemistry, and served as carriers of IR780 photothermal agent. With an excellent IR780 loading content of up to ca. 25%, these molecular nanovehicles still maintained their molecular morphologies and did not aggregate in cell culture medium. Among three MBB, the rodlike one exhibited best performance in cell uptake in the 2D and also in spheroid penetration in 3D cell culture. Furthermore, this rodlike system had preferential accumulation in tumor in vivo and excellent effect on photothermal cancer therapy which effectively inhibited tumor growth. These results demonstrated an important role of nanoparticle shape on bio-behaviors and the unimolecular micelle could be a promising nanovehicle with precisely defined structure for biomedicine applications.


Subject(s)
Hyperthermia, Induced , Micelles , Neoplasms/therapy , Phototherapy , Animals , Cell Survival/drug effects , Endocytosis/drug effects , Female , Humans , Indoles/pharmacology , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Neoplasms/pathology , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Static Electricity , Tissue Distribution/drug effects , Tumor Burden/drug effects
8.
Adv Healthc Mater ; 7(11): e1800121, 2018 06.
Article in English | MEDLINE | ID: mdl-29582583

ABSTRACT

Autophagy is a homeostatic lysosome-dependent metabolic process to eliminate damaged or dysfunctional cellular organelles, which is closely associated with tumor progression. Indocyanine green (ICG) can convert NIR light energy to localized heat for cancer cell and tissue ablation. However, the effect of autophagy modulation on ICG-mediated photothermal therapy remains unknown. In this study, it is found that primaquine (PQ) suppresses autophagy flux at a late stage through the impeding fusion of the autophagosome with the lysosome to form an autophagolysosome, leading to cell apoptosis or necrosis. This autophagosome-lysosome fusion inhibitory effect and the autophagosome accumulation are more evident in the photothermal therapy combined with autophagy inhibition. Motivated by this notable effect, a cascade-targeting nanocapsule (HCP) is constructed using an organic solvent-free strategy to coencapsulate PQ and ICG. By targeting the cluster designation 44 molecule and sequentially enhancing the cell-penetrating peptide-mediated endocytosis, the codelivery of PQ/ICG by HCP achieves selective recognition and reinforces the internalization by MCF-7 cells to exert a synergistic therapeutic effect on MCF-7 cells both in vitro and in vivo. The HCP system for the photothermal and autophagy inhibition combination therapy represents a novel strategy for the treatment of breast cancer.


Subject(s)
Autophagy/drug effects , Breast Neoplasms/therapy , Cell-Penetrating Peptides , Hyperthermia, Induced , Nanocapsules , Phototherapy , Primaquine , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacokinetics , Cell-Penetrating Peptides/pharmacology , Endocytosis/drug effects , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Nanocapsules/chemistry , Nanocapsules/therapeutic use , Primaquine/chemistry , Primaquine/pharmacokinetics , Primaquine/pharmacology , Xenograft Model Antitumor Assays
9.
J Ethnopharmacol ; 217: 118-125, 2018 May 10.
Article in English | MEDLINE | ID: mdl-29421593

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Jiang-Xian HuGan (JXHG) formulated by five natural products including Freshwater clam (Corbicula fluminea), Curcuma longa L., Ligustrum lucidum, Eclipta prostrata (L.) L. and Paeonia lactiflora Pall., has exhibited a great hepatoprotective effect. AIM OF THIS STUDY: We investigated the effect of JXHG on concanavalin A (ConA)-induced acute live injury in mice, and to elucidate its underlying molecular mechanisms. MATERIALS AND METHODS: Jiangkanling Capsule (900 mg/kg), low-dose JXHG (LJXHG, 700 mg/kg), high-dose JXHG (HJXHG, 1400 mg/kg) were administered to mice by oral gavage daily for 20 days prior to a single intravenous injection of ConA (20 mg/kg). Liver injury was evaluated by measuring the serum levels of enzymes and cytokines as well as liver histological analysis. We also measured the hepatic expression of cytokines at mRNA levels and the proteins related to NF-κB and Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) signaling pathways. RESULT: Our results showed that JXHG pretreatment significantly alleviated ConA-induced live injury as evidenced by decreased serum levels of glutamic-pyruvic transaminase (ALT) and glutamic oxalacetic transaminase (AST), and reduced hepatocyte apoptosis and mortality. Furthermore, JXHG was able to significantly reduce the serum levels of proinflammatory cytokines, down-regulate the mRNA expression of interleukin-6 (IL-6) and interferon-γ (IFN-γ), and up-regulate IL-10 as well as superoxide-dimutase-1 (SOD1), glutathione reductase (GSR) and Glutathione peroxidase 2 (GPX2) mRNA in the liver tissues after Con A injection. In addition, JXHG pretreatment dramatically suppressed the phosphorylation of NF-κB p65 (p65), increased Nrf2 expression, and decreased the expression ratio of cleaved caspase-3/caspase-3 in liver tissues. CONCLUSION: These results suggest that JXHG protects against ConA-induced acute live injury through inhibiting NF-κB mediated inflammatory pathway and promoting Nrf2 mediated anti-oxidative stress signaling pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Chemical and Drug Induced Liver Injury/prevention & control , Concanavalin A , Drugs, Chinese Herbal/pharmacology , Liver/drug effects , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Animals , Biomarkers/metabolism , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Curcumin/pharmacology , Cytokines/metabolism , Disease Models, Animal , Enzymes/metabolism , Inflammation Mediators/metabolism , Liver/metabolism , Liver/pathology , Male , Mice, Inbred BALB C , Oleanolic Acid/pharmacology , Oxidative Stress/drug effects , Signal Transduction/drug effects
10.
J Nat Med ; 72(1): 145-154, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28936800

ABSTRACT

Liver fibrosis is considered to be a result of chronic liver pathological changes, and hepatic stellate cells (HSCs) play an important role during this process. Evodiamine, an indole alkaloid derived from Evodia rutaecarpa, exhibits pharmacological activities. This study focused on the effects of evodiamine on carbon tetrachloride (CCl4)-induced liver fibrosis in rats and HSCs in vitro via the TGF-ß1/Smad signaling pathway. A liver fibrosis rat model was established by the intraperitoneal injection of CCl4 (3 ml/kg, 30% in olive oil). Evodiamine (15 and 25 mg/kg) was administered orally for 8 weeks. HSCs were treated with different evodiamine concentrations. The results indicated that evodiamine could improve the histopathological abnormalities in liver tissues and decrease the level of aspartate aminotransferase (AST), alanine aminotransferase (ALT), hydroxyproline, and total bilirubin (TBIL). Concentrations of IL-6, tumor necrosis factor-α (TNF-α), collagen-I (COL-I), and collagen-III (COL-III) were reduced by evodiamine. Western blotting and real-time PCR showed that protein expression of transforming growth factor-ß (TGF-ß1), p-Smad 2/3 (phosphorylation of Smad 2/3), and smooth muscle alpha-actin (α-SMA) as well as mRNA expression of TGF-ß1 and α-SMA in liver tissues were downregulated by evodiamine. The cell proliferation, production of hydroxyproline, and the protein expression of TGF-ß1, p-Smad 2/3, and α-SMA in HSCs were dose-dependently reduced by evodiamine. Collectively, evodiamine had an antifibrosis effect in CCl4-induced liver fibrosis, and reduced HSCs proliferation and collagen metabolism in vitro. The major mechanism was downregulation of relative expression of TGF-ß1, p-Smad 2/3, and α-SMA.


Subject(s)
Liver Cirrhosis/drug therapy , Quinazolines/therapeutic use , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Disease Models, Animal , Liver Cirrhosis/pathology , Male , Quinazolines/administration & dosage , Quinazolines/pharmacology , Rats , Rats, Wistar
11.
Med. oral patol. oral cir. bucal (Internet) ; 21(3): e335-e340, mayo 2016. tab
Article in English | IBECS | ID: ibc-152713

ABSTRACT

BACKGROUND: The aim of this study was to assess the serum zinc levels in patients with common oral mucosal diseases by comparing these to healthy controls. MATERIAL AND METHODS: A total of 368 patients, which consisted of 156 recurrent aphthous stomatitis (RAS) patients, 57 oral lichen planus (OLP) patients, 55 burning mouth syndrome (BMS) patients, 54 atrophic glossitis (AG) patients, 46 xerostomia patients, and 115 sex-and age-matched healthy control subjects were enrolled in this study. Serum zinc levels were measured in all participants. Statistical analysis was performed using a one-way ANOVA, t-test, and Chi-square test. RESULTS: The mean serum zinc level in the healthy control group was significantly higher than the levels of all other groups (p < 0.001). No individual in the healthy control group had a serum zinc level less than the minimum normal value. However, up to 24.7% (13/54) of patients with AG presented with zinc deficiency, while 21.2% (33/156) of patients with RAS, 16.4% (9/55) of patients with BMS, 15.2% (7/46) of patients with xerostomia, and 14.0% (8/57) of patients with OLP were zinc deficient. Altogether, the zinc deficiency rate was 19.02% (70/368) in the oral mucosal diseases (OMD) group (all patients with OMD). The difference between the OMD and healthy control group was significant (p< 0.001). Gender differences in serum zinc levels were also present, although not statistically significant. CONCLUSIONS: Zinc deficiency may be involved in the pathogenesis of common oral mucosal diseases. Zinc supplementation may be a useful treatment for oral mucosal diseases, but this requires further investigation; the optimal serum level of zinc, for the prevention and treatment of oral mucosal diseases, remains to be determined


Subject(s)
Humans , Stomatognathic Diseases/physiopathology , Zinc/blood , Zinc Deficiency , Mouth Mucosa/physiopathology , Dietary Supplements
12.
BMC Microbiol ; 15: 274, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26630969

ABSTRACT

BACKGROUND: Small-molecule compounds that inhibit human immunodeficiency virus type 1 (HIV-1) infection can be used not only as drug candidates, but also as reagents to dissect the life cycle of the virus. Thus, it is desirable to have an arsenal of such compounds that inhibit HIV-1 infection by various mechanisms. Until now, only a few small-molecule compounds that inhibit nuclear entry of viral DNA have been documented. RESULTS: We identified a novel, small-molecule compound, SJP-L-5, that inhibits HIV-1 infection. SJP-L-5 is a nitrogen-containing, biphenyl compound whose synthesis was based on the dibenzocyclooctadiene lignan gomisin M2, an anti-HIV bioactive compound isolated from Schisandra micrantha A. C. Smith. SJP-L-5 displayed relatively low cytotoxicity (50% cytoxicity concentrations were greater than 200 µg/ml) and high antiviral activity against a variety of HIV strains (50% effective concentrations (EC50)) of HIV-1 laboratory-adapted strains ranged from 0.16-0.97 µg/ml; EC50s of primary isolates ranged from 1.96-5.33 µg/ml). Analyses of the viral DNA synthesis indicated that SJP-L-5 specifically blocks the entry of the HIV-1 pre-integration complex (PIC) into the nucleus. Further results implicated that SJP-L-5 inhibits the disassembly of HIV-1 particulate capsid in the cytoplasm of the infected cells. CONCLUSIONS: SJP-L-5 is a novel small-molecule compound that inhibits HIV-1 nuclear entry by blocking the disassembly of the viral core.


Subject(s)
Anti-HIV Agents/pharmacology , DNA, Viral/metabolism , HIV-1/drug effects , HIV-1/physiology , Virus Integration/drug effects , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/isolation & purification , Anti-HIV Agents/toxicity , Cell Line , Cell Survival/drug effects , Humans , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Schisandra/chemistry
13.
Mol Neurobiol ; 51(2): 685-95, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24915968

ABSTRACT

Diabetes mellitus substantially increases the risk of stroke and enhances brain's vulnerability to ischemia insult. Electroacupuncture (EA) pretreatment was proved to induce cerebral ischemic tolerance in normal stroke models. Whether EA could attenuate cerebral ischemia injury in diabetic mice and the possible underlying mechanism are still unrevealed. Male C57BL/6 mice were subjected to streptozotocin (STZ) for diabetic models. After inducing focal cerebral ischemia model, the levels of plasma and cerebral adiponectin (APN) were measured as well as the expression of cerebral adiponectin receptor 1 (AdipoR1) and 2 (AdipoR2). The neurobehavioral score, infarction volume, and cellular apoptosis were evaluated with or without AdipoR1 short interfering RNA (siRNA). The role of phosphorylation of glycogen synthesis kinase 3 beta (GSK-3ß) at Ser-9 in the EA pretreatment was also assessed. EA pretreatment increased both plasma and cerebral APN levels and enhanced neuronal AdipoR1 in diabetic mice. In addition, EA reduced infarct size, improved neurological outcomes, and inhibited cell apoptosis after reperfusion. These beneficial effects were reversed by AdipoR1 knockdown. Furthermore, EA increased GSK-3ß phosphorylation (p-GSK-3ß) in the ipsilateral penumbra. Augmented p-GSK-3ß induced neuroprotective effects similar to those of EA pretreatment. In contrast, dampened p-GSK-3ß could reverse the neuroprotective effects of EA. In addition, the increase in p-GSK-3ß by EA was abolished by AdipoR1 knockdown. We conclude that EA pretreatment increases the production of APN, which induce protective effects against cerebral ischemia-reperfusion injury through neuronal AdipoR1-mediated phosphorylation of GSK-3ß in diabetic mice.


Subject(s)
Brain Ischemia/metabolism , Diabetes Mellitus, Experimental/metabolism , Electroacupuncture/methods , Glycogen Synthase Kinase 3/metabolism , Receptors, Adiponectin/physiology , Reperfusion Injury/metabolism , Animals , Brain Ischemia/prevention & control , Diabetes Mellitus, Experimental/therapy , Glycogen Synthase Kinase 3 beta , Male , Mice , Mice, Inbred C57BL , Phosphorylation/physiology , Reperfusion Injury/prevention & control
14.
Brain Res ; 1573: 84-91, 2014 Jul 21.
Article in English | MEDLINE | ID: mdl-24854123

ABSTRACT

We investigated the protective effect of electroacupuncture (EA) on cerebral ischemic injury in diabetic mice, and explored the role of NADPH oxidase-mediated oxidative stress. Male C57BL/6 mice were injected streptozotocin to induce diabetes. The mice were pretreated with EA at acupoint "Baihui" for 30 min. Two hours after the end of EA pretreatment, focal cerebral ischemia was induced following 24h reperfusion. The neurobehavioral scores and infarction volumes, malondialdehyde (MDA), reactive oxygen species (ROS), and activation of NADPH oxidase were determined in the presence or absence of the NADPH oxidase inhibitor apocynin or activator tetrabromocinnamic acid (TBCA). EA pretreatment reduced infarct size and improved neurological outcomes 24h after reperfusion in the diabetic mice. EA also decreased cerebral MDA and ROS levels compared with the control group, and inhibited the NADPH oxidase activation. The beneficial effects were abolished by TBCA while pretreatment with apocynin mimicked the neuroprotective and anti-oxidative effects of EA. Our results demonstrated that EA attenuated cerebral ischemic injury by inhibiting NAPDH oxidase-mediated oxidative damage in diabetic mice. These results suggest a novel mechanism of EA pretreatment-induced tolerance in diabetic cerebral ischemia.


Subject(s)
Brain Ischemia/therapy , Diabetes Mellitus, Experimental/complications , Electroacupuncture/methods , NADPH Oxidases/metabolism , Oxidative Stress/physiology , Acetophenones/pharmacology , Animals , Brain/drug effects , Brain/enzymology , Brain/pathology , Brain Ischemia/complications , Brain Ischemia/enzymology , Brain Ischemia/pathology , Cinnamates/pharmacology , Enzyme Activators/pharmacology , Male , Malondialdehyde/metabolism , Mice, Inbred C57BL , NADPH Oxidases/antagonists & inhibitors , Oxidative Stress/drug effects , Random Allocation , Reactive Oxygen Species/metabolism , Reperfusion Injury/complications , Reperfusion Injury/enzymology , Reperfusion Injury/prevention & control
15.
BMC Complement Altern Med ; 14: 37, 2014 Jan 22.
Article in English | MEDLINE | ID: mdl-24447795

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is a severe neurodegenerative disease for which there is currently no effective treatment. The purpose of this study was to investigate whether repeated electroacupuncture (EA) stimulation would improve cognitive function and the pathological features of AD in amyloid precursor protein (APP)/presenilin 1 (PS1) double transgenic mice. METHODS: Cognitive function of APP/PS1 double transgenic mice was assessed using the Morris water maze test before and after EA treatment. Levels of amyloid ß-peptide (Aß) deposits in the hippocampus and cortex were evaluated by immunofluorescence, western blot and enzyme-linked immunosorbent assay. Expression of brain-derived neurotrophic factor (BDNF) was also examined by immunofluorescence and western blot. The neurogenesis was labeled by the DNA marker bromodeoxyuridine. RESULTS: EA stimulation significantly ameliorated the learning and memory deficits of AD mice by shortening escape latency and increasing the time spent in the target zone during the probe test. Additionally, decreased Aß deposits and increased BDNF expression and neurogenesis in the hippocampus and cortex of EA-treated AD mice were detected. The same change was detected in wild-type mice after EA treatment compared with wild-type mice without EA treatment. CONCLUSIONS: Repeated EA stimulation may improve cognitive function, attenuate Aß deposits, up-regulate the expression of BDNF and promote neurogenesis in the APP/PS1 double transgenic mice. This suggests that EA may be a promising treatment for AD.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Brain/metabolism , Cognition Disorders/therapy , Electroacupuncture , Memory Disorders/therapy , Neurogenesis , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cognition , Cognition Disorders/etiology , Cognition Disorders/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Maze Learning , Memory Disorders/etiology , Memory Disorders/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1/genetics , Presenilin-1/metabolism
16.
Mol Neurobiol ; 50(2): 305-13, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24390566

ABSTRACT

Electroacupuncture (EA) has demonstrated therapeutic potential for the treatment of Alzheimer's disease (AD). A previous study reported that N-myc downstream-regulated gene 2 (NDRG2) was upregulated in the brain of patients with AD. In the present study, we investigated the effects of repeated EA administration on reference memory impairment and the role of NDRG2 in an amyloid precursor protein (APP)/presenilin-1 (PS1) double transgenic mouse model. Age-matched wild-type and transgenic mice were treated with EA (once per day for 30 min) for 4 weeks (four courses of 5 days EA administration and 2 days rest) beginning at 10 months of age. At seven and ten postnatal months of age and following a 4-week EA treatment regime, mice received training in the Morris water maze (MWM) and a probe test. Brain tissue was analyzed via Western blot and double-label immunofluorescence. Beginning at 7 months of age, APP/PS1 mice began to exhibit deficits in reference memory in the MWM test, an impairment associated with upregulation of glial fibrillary acidic protein (GFAP) and NDRG2. Four weeks of EA administration significantly ameliorated cognitive impairments and suppressed GFAP and NDRG2 upregulation. In conclusion, our findings demonstrated that EA administration can alleviate reference memory deficits and suppress NDRG2 upregulation in an AD transgenic mouse model. This study provides supportive evidence for EA as an effective therapeutic intervention for AD, as well as NDRG2 as a novel target for AD treatment.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Protein Precursor/metabolism , Astrocytes/metabolism , Electroacupuncture , Memory Disorders/therapy , Presenilin-1/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Behavior, Animal/physiology , Disease Models, Animal , Memory/physiology , Mice , Mice, Transgenic
17.
Biochem Pharmacol ; 86(12): 1762-74, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24144633

ABSTRACT

Elevated levels of the glycolysis metabolite methylglyoxal (MG) have been implicated in impaired leukocyte-endothelial interactions and vascular complications in diabetes, putative mechanisms of which remain elusive. Uncoupling of endothelial nitric oxide synthase (eNOS) was shown to be involved in endothelial dysfunction in diabetes. Whether MG contributes to these effects has not been elucidated. By using intravital microscopy in vivo, we demonstrate that MG-triggered reduction in leukocyte rolling velocity and increases in rolling flux, adhesion, emigration and microvascular permeability were significantly abated by scavenging reactive oxygen species (ROS). In murine cremaster muscle, MG treatment reduced tetrahydrobiopterin (BH4)/total biopterin ratio, increased arginase expression and stimulated ROS and superoxide production. The latter was significantly blunted by ROS scavengers Tempol (300µM) or MnTBAP (300µM), by BH4 supplementation (100µM) or by NOS inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME; 20µM). In these tissues and cultured murine and human primary endothelial cells, MG increased eNOS monomerization and decreased BH4/total biopterin ratio, effects that were significantly mitigated by supplementation of BH4 or its precursor sepiapterin but not by L-NAME or tetrahydroneopterin, indicative of MG-triggered eNOS uncoupling. MG treatment further decreased the expression of guanosine triphosphate cyclohydrolase I in murine primary endothelial cells. MG-induced leukocyte recruitment was significantly attenuated by supplementation of BH4 or sepiapterin or suppression of superoxide by L-NAME confirming the role of eNOS uncoupling in MG-elicited leukocyte recruitment. Together, our study uncovers eNOS uncoupling as a pivotal mechanism in MG-induced oxidative stress, microvascular hyperpermeability and leukocyte recruitment in vivo.


Subject(s)
Capillary Permeability/drug effects , Leukocytes/cytology , Nitric Oxide Synthase Type III/metabolism , Pyruvaldehyde/pharmacology , Animals , Biopterins/analogs & derivatives , Biopterins/metabolism , Leukocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism
18.
Cardiovasc Diabetol ; 12: 134, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-24050620

ABSTRACT

BACKGROUND: Increased levels of the sugar metabolite methylglyoxal (MG) in vivo were shown to participate in the pathophysiology of vascular complications in diabetes. Alterations of endothelial nitric oxide synthase (eNOS) activity by hypophosphorylation of the enzyme and enhanced monomerization are found in the diabetic milieu, and the regulation of this still remains undefined. Using various pharmacological approaches, we elucidate putative mechanisms by which MG modulates eNOS-associated functions of MG-stimulated superoxide O2•⁻ production, phosphorylation status and eNOS uncoupling in EA.hy926 human endothelial cells. METHODS: In cultured EA.hy926 endothelial cells, the effects of MG treatment, tetrahydrobiopterin (BH4; 100 µM) and sepiapterin (20 µM) supplementation, NOS inhibition by N(G)-nitro-L-arginine methyl ester (L-NAME; 50 µM), and inhibition of peroxynitrite (ONOO⁻) formation (300 µM Tempol plus 50 µM L-NAME) on eNOS dimer/monomer ratios, Ser-1177 eNOS phosphorylation and 3-nitrotyrosine (3NT) abundance were quantified using immunoblotting. O2•⁻-dependent fluorescence was determined using a commercially available kit and tissue biopterin levels were measured by fluorometric HPLC analysis. RESULTS: In EA.hy926 cells, MG treatment significantly enhanced O2•⁻ generation and 3NT expression and reduced Ser-1177 eNOS phosphorylation, eNOS dimer/monomer ratio and cellular biopterin levels indicative of eNOS uncoupling. These effects were significantly mitigated by administration of BH4, sepiapterin and suppression of ONOO⁻ formation. L-NAME treatment significantly blunted eNOS-derived O2•⁻ generation but did not modify eNOS phosphorylation or monomerization. CONCLUSION: MG triggers eNOS uncoupling and hypophosphorylation in EA.hy926 endothelial cells associated with O2•⁻ generation and biopterin depletion. The observed effects of the glycolysis metabolite MG presumably account, at least in part, for endothelial dysfunction in diabetes.


Subject(s)
Endothelial Cells/drug effects , Nitric Oxide Synthase Type III/metabolism , Pyruvaldehyde/toxicity , Antioxidants/pharmacology , Biopterins/metabolism , Cell Line , Dose-Response Relationship, Drug , Endothelial Cells/enzymology , Enzyme Inhibitors/pharmacology , Humans , Nitric Oxide Synthase Type III/antagonists & inhibitors , Peroxynitrous Acid/metabolism , Phosphorylation , Protein Multimerization , Superoxides/metabolism , Time Factors , Tyrosine/analogs & derivatives , Tyrosine/metabolism
19.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 29(11): 1012-7, 2009 Nov.
Article in Chinese | MEDLINE | ID: mdl-20329614

ABSTRACT

OBJECTIVE: To explore the mechanism of action of tanshinone II A for liver protection in hepatic fibrotic mice by observing its effects on signaling pathway of insulin-like growth factor binding protein 7 (IGFBP7) and TGFbeta1/Smad3. METHODS: Hepatic fibrosis model was induced by intraperitoneal injection of thioacetamide (TAA). Thirty-six male Kunming mice were divided into five groups: the normal control group (N), the 4-week model group (A), the 4-week tanshinone II A prevented group (B), the 6-week model group (C) and the 3-week tanshinone II A treated group (D). Changes of serum levels of alanine aminotransferase (ALT) and lactate dehydrogenase (LDH), histopathology of liver (HE staining), area density of collagen in liver (Masson staining), expressions of alpha-smooth muscle actin (alpha-SMA), collagen I , fibronectin (FN), transforming growth factor-beta1 (TGF-beta1), Smad3 and IGFBP7 in liver (by immunohiStochemistry), liver content of FN, Smad3 and IGFBP7 (by Western blot), and the hepatocyte apoptosis (by TUNEL) were observed. RESULTS: The serum levels of ALT and LDH, the expressions of alpha-SMA, collagen I , TGF-beta1 in liver, expressions and contents of FN, Smad3 and IGFBP7 in liver were significantly lower; the liver damage and the hepatic apoptosis index were lesser in Group B than in Group A, also in Group D than in Group C, respectively, all showing statistical significance (P < 0.05). CONCLUSION: Tanshinone II A could improve liver function, inhibit the activation of hepatic stellate cells, reduce the production of extracellular matrix, and protect the hepatocytes, and its of mechanisms of actions might be related with blocking TGF-beta1/Smad3 signaling pathway and down-regulating the expression of IGFBP7 in liver.


Subject(s)
Abietanes/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver Cirrhosis, Experimental/metabolism , Animals , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Insulin-Like Growth Factor Binding Proteins/metabolism , Male , Mice , Mice, Inbred Strains , Signal Transduction , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL