Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Redox Biol ; 36: 101642, 2020 09.
Article in English | MEDLINE | ID: mdl-32863238

ABSTRACT

Normal high-density lipoprotein (nHDL) in normal, healthy subjects is able to promote angiogenesis, but the mechanism remains incompletely understood. HDL from patients with coronary artery disease may undergo a variety of oxidative modifications, rendering it dysfunctional; whether the angiogenic effect is mitigated by such dysfunctional HDL (dHDL) is unknown. We hypothesized that dHDL compromises angiogenesis. The angiogenic effects of nHDL and dHDL were assessed using endothelial cell culture, endothelial sprouts from cardiac tissue from C57BL/6 mice, zebrafish model for vascular growth and a model of impaired vascular growth in hypercholesterolemic low-density lipoprotein receptor null(LDLr-/-)mice. MiRNA microarray and proteomic analyses were used to determine the mechanisms. Lipid hydroperoxides were greater in dHDL than in nHDL. While nHDL stimulated angiogenesis, dHDL attenuated these responses. Protein and miRNA profiles in endothelial cells differed between nHDL and dHDL treatments. Moreover, nHDL suppressed miR-24-3p expression to increase vinculin expression resulting in nitric oxide (NO) production, whereas dHDL delivered miR-24-3p to inhibit vinculin expression leading to superoxide anion (O2•-) generation via scavenger receptor class B type 1. Vinculin was required for endothelial nitric oxide synthase (eNOS) expression and activation and modulated the PI3K/AKT/eNOS and ERK1/2 signaling pathways to regulate nHDL- and VEGF-induced angiogenesis. Vinculin overexpression or miR-24-3p inhibition reversed dHDL-impaired angiogenesis. The expressions of vinculin and eNOS and angiogenesis were decreased, but the expression of miR-24-3p and lipid hydroperoxides in HDL were increased in the ischemic lower limbs of hypercholesterolemic LDLr-/- mice. Overexpression of vinculin or miR-24-3p antagomir restored the impaired-angiogenesis in ischemic hypercholesterolemic LDLr-/- mice. Collectively, nHDL stimulated vinculin and eNOS expression to increase NO production by suppressing miR-24-3p to induce angiogenesis, whereas dHDL inhibited vinculin and eNOS expression to enhance O2•- generation by delivering miR-24-3p to impair angiogenesis, and that vinculin and miR-24-3p may be therapeutic targets for dHDL-impaired angiogenesis.


Subject(s)
Coronary Artery Disease , MicroRNAs , Animals , Coronary Artery Disease/drug therapy , Coronary Artery Disease/genetics , Endothelial Cells , Healthy Volunteers , Humans , Lipoproteins, HDL , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Neovascularization, Physiologic , Nitric Oxide Synthase Type III/genetics , Phosphatidylinositol 3-Kinases , Proteomics , Zebrafish
3.
PLoS One ; 10(6): e0127775, 2015.
Article in English | MEDLINE | ID: mdl-26068440

ABSTRACT

Obstructive sleep apnea (OSA) is much more prevalent in older people than in middle-aged or young populations, and has been associated with cardiovascular disease. Continuous positive airway pressure (CPAP) is the first-line therapy for OSA, but its long-term clinical benefit in the elderly is unclear. Here, we carried out a prospective cohort study to explore the survival rate and incidence of cardiovascular events in elderly patients with moderate to severe OSA who did or did not receive CPAP treatment. The study included 130 patients (104 male, 26 female; mean age: 77.8 ± 6.2 years) who were followed up for a mean of 5 ± 2.54 years (range, 1-8 years). Thirty-six patients received CPAP and 88 had no CPAP. The results showed that mortality in the untreated group (21.6%) was significantly higher than in the CPAP group (5.6%). Kaplan-Meier survival analysis showed that the survival rate in the CPAP group was 94.4%, which was markedly higher than the rate of 78.4% in the untreated group. The incidence of cardiovascular events was 13.9% in the CPAP group and 55.7% in the untreated group. The present study provides evidence that CPAP can reduce mortality in older patients with moderate to severe OSA, and lead to a good long-term prognosis. The study also indicates that death in older OSA patients is associated with cardiovascular disease and diabetes.


Subject(s)
Continuous Positive Airway Pressure , Sleep Apnea, Obstructive/therapy , Aged , Aged, 80 and over , Cardiovascular Diseases/etiology , Cohort Studies , Female , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Male , Multivariate Analysis , Prospective Studies , Severity of Illness Index , Sleep Apnea, Obstructive/complications , Sleep Apnea, Obstructive/mortality
4.
BMC Complement Altern Med ; 15: 137, 2015 Apr 29.
Article in English | MEDLINE | ID: mdl-25925762

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is the second most common neurodegenerative disease, affecting 2% of the population aged over 65 years old. Mitochondrial defects and oxidative stress actively participate in degeneration of dopaminergic (DA) neurons in PD. Paeonolum, a main component isolated from Moutan cortex, has potent antioxidant ability. Here, we have examined the effects of paeonolum against MPP(+)-induced neurotoxicity in zebrafish and PC12 cells. METHODS: The overall viability and neurodegeneration of DA neurons was assessed in ETvmat2:green fluorescent protein (GFP) transgenic zebrafish, in which most monoaminergic neurons are labeled by GFP. Damage to PC12 cells was measured using a cell viability assay and assessment of nuclear morphology. Intracellular reactive oxygen species (ROS) and the level of total GSH were assessed. The mitochondrial cell death pathway including mitochondrial membrane potential, cytochrome C release and caspase-3 activity were also examined in PC12 cells. RESULTS: Paeonolum protected against MPP(+)-induced DA neurodegeneration and locomotor dysfunction in zebrafish in a concentration-dependent manner. Similar neuroprotection was replicated in the PC12 cellular model of MPP(+) toxicity. Paeonolum attenuated MPP(+)-induced intracellular ROS accumulation and restored the level of total GSH in PC12 cells. Furthermore, paeonolum significantly inhibited the mitochondrial cell death pathway induced by MPP(+). CONCLUSIONS: Collectively, the present study demonstrates that paeonolum protects zebrafish and PC12 cells against MPP(+)-induced neurotoxicity.


Subject(s)
Antioxidants/therapeutic use , Dopaminergic Neurons/drug effects , Neuroprotective Agents/therapeutic use , Paeonia/chemistry , Parkinson Disease/prevention & control , Phytotherapy , Plant Extracts/therapeutic use , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Caspase 3/metabolism , Cell Death/drug effects , Cytochromes c/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Humans , MPTP Poisoning/metabolism , MPTP Poisoning/prevention & control , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Neuroprotective Agents/pharmacology , Neurotoxins/adverse effects , Oxidative Stress/drug effects , PC12 Cells , Parkinson Disease/metabolism , Plant Extracts/pharmacology , Rats , Reactive Oxygen Species/metabolism , Zebrafish
5.
Mar Drugs ; 12(6): 3307-22, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24886868

ABSTRACT

High-throughput behavior-based screen in zebrafish is a powerful approach for the discovery of novel neuroactive small molecules for treatment of nervous system diseases such as epilepsy. To identify neuroactive small molecules, we first screened 36 compounds (1-36) derived from marine natural products xyloketals and marine isoprenyl phenyl ether obtained from the mangrove fungus. Compound 1 demonstrated the most potent inhibition on the locomotor activity in larval zebrafish. Compounds 37-42 were further synthesized and their potential anti-epilepsy action was then examined in a PTZ-induced epilepsy model in zebrafish. Compound 1 and compounds 39, 40 and 41 could significantly attenuate PTZ-induced locomotor hyperactivity and elevation of c-fos mRNA in larval zebrafish. Compound 40 showed the most potent inhibitory action against PTZ-induced hyperactivity. The structure-activity analysis showed that the OH group at 12-position played a critical role and the substituents at the 13-position were well tolerated in the inhibitory activity of xyloketal derivatives. Thus, these derivatives may provide some novel drug candidates for the treatment of epilepsy.


Subject(s)
Anticonvulsants/pharmacology , Epilepsy/drug therapy , Phenyl Ethers/pharmacology , Pyrans/pharmacology , Animals , Anticonvulsants/chemistry , Anticonvulsants/isolation & purification , Behavior, Animal/drug effects , Biological Products/chemistry , Biological Products/isolation & purification , Biological Products/pharmacology , Disease Models, Animal , Fungi/chemistry , High-Throughput Screening Assays/methods , Larva , Motor Activity/drug effects , Oceans and Seas , Pentylenetetrazole , Phenyl Ethers/chemistry , Phenyl Ethers/isolation & purification , Proto-Oncogene Proteins c-fos/genetics , Pyrans/chemistry , Pyrans/isolation & purification , RNA, Messenger/metabolism , Structure-Activity Relationship , Zebrafish
6.
Eur J Pharmacol ; 732: 76-85, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24690262

ABSTRACT

Aß40-induced vascular dysfunction has been implicated in the pathogenesis of Alzheimer׳s disease (AD). In the present study, we investigated the possible protective effects of puerarin against Aß40-induced vascular damage and impairment to angiogenesis in transgenic TG (fli1:EGFP) zebrafish and human endothelial cells. Aß40 peptides at 5µM caused an obvious reduction of vessel branches in the subintestinal vein basket, induced NADPH oxidase-derived reactive oxygen species and impaired vascular endothelial growth factor (VEGF)-dependent angiogenesis. Pretreatment with puerarin attenuated Aß40-induced vessel reduction and impairment to angiogenesis in a dose-dependent manner. In addition, Aß40 decreased VEGF-dependent phosphorylation of Akt and eNOS, whereas puerarin treatment attenuated these detrimental effects. Furthermore, the restoration of Aß40-induced-angiogenesis impairment by puerarin was abolished by either the PI3 kinase inhibitor LY294002 (10µM) or eNOS inhibitor L-NAME. The present study suggests that puerarin exerts its protective action probably through reduction of NADPH oxidase-derived reactive oxygen species overproduction and activation of the PI3K/Akt/eNOS pathways.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/toxicity , Endothelial Cells/drug effects , Isoflavones/pharmacology , Neuroprotective Agents/pharmacology , Vascular Diseases/chemically induced , Vascular Diseases/prevention & control , Animals , Humans , Neovascularization, Physiologic/drug effects , Reactive Oxygen Species/metabolism , Respiratory Burst/drug effects , Vascular Diseases/pathology , Zebrafish
7.
PLoS One ; 8(7): e68106, 2013.
Article in English | MEDLINE | ID: mdl-23874513

ABSTRACT

Mutations in the TARDBP gene, which encodes the Tar DNA binding protein, have been shown to causes of both familial amyotrophic lateral sclerosis (FALS) and sporadic ALS (SALS). Recently, several novel TARDBP exon 6 mutants have been reported in patients with ALS in Europe and America but not in Asia. To further examine the spectrum and frequency of TARDBP exon 6 mutations, we investigated their frequency in ethnic Chinese patients with sporadic ALS. TARDBP exon 6 was screened by direct sequencing in 207 non-SOD1 SALS patients and 230 unrelated healthy controls but no mutations were identified. Our data indicate that exon 6 mutations in TARDBP are not a common cause of SALS in Han Chinese population from Southern Mainland China.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Asian People/genetics , DNA-Binding Proteins/genetics , Exons , Mutation , Adult , China , Female , Genetic Association Studies , Humans , Male , Middle Aged
8.
PLoS One ; 8(5): e64720, 2013.
Article in English | MEDLINE | ID: mdl-23724084

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder mainly affecting motor neurons. Mutations in superoxide dismutase-1 (SOD-1) account for about 20% of familial ALS patients. A robust supply of motoneurons carrying the mutated gene would help understand the causes of motoneuron death and develop new therapeutics for the disease. Here, we established induced pluripotent stem (iPS) cell lines from SOD1G93A mice and compared their potency in motoneuron generation with normal iPS cells and mouse embryonic stem cells (E14). Our results showed that iPS cells derived from SOD1G93A mice possessed the similar potency in neuronal differentiation to normal iPS cells and E14 cells and can be efficiently driven to motoneuron-like phenotype. These cells exhibited typical neuronal morphology, expressed key motoneuron markers, including ChAT and HB9, and generated repetitive trains of action potentials. Furthermore, these neurons highly expressed human SOD-1 and exhibited shorter neurites compared to controls. The present study provides evidence that ALS-iPS cells can be used as disease models in high-throughput screening and mechanistic studies due to their ability to efficiently differentiate into specific neuronal subtypes.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Motor Neurons/cytology , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Line , Fibroblasts/cytology , Fibroblasts/metabolism , Green Fluorescent Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Promoter Regions, Genetic/genetics , Superoxide Dismutase-1 , Tail , Transduction, Genetic , Tubulin/metabolism
9.
Mol Divers ; 17(3): 515-24, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23703159

ABSTRACT

The effects of Salvianolic acid A (Sal A) on the treatment of Alzheimer's disease (AD) were investigated. Sal A significantly inhibits amyloid beta [Formula: see text] self-aggregation and disaggregates pre-formed [Formula: see text] fibrils, reduces metal-induced [Formula: see text] aggregation through chelating metal ions, and blocks the formation of reactive oxygen species (ROS) in SH-SY5Y cells. Sal A protects cells against [Formula: see text]-induced toxicity. Furthermore, Sal A, possibly because of the effects of decreasing toxicity effects of [Formula: see text] species, alleviates [Formula: see text]-induced paralysis in transgenic Caenorhabditis elegans. Circular dichroism (CD) experiments and Molecular dynamic (MD) simulations demonstrate that Sal A inhibits [Formula: see text] self-aggregation through binding to the C-terminus of [Formula: see text], and therefore stabilizing the [Formula: see text]-helical conformations. Altogether, our data show that Sal A, as the multifunctional agent, is likely to be promising therapeutics for AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/drug effects , Caffeic Acids/pharmacology , Lactates/pharmacology , Plaque, Amyloid/drug therapy , Serum Amyloid A Protein/drug effects , Animals , Caenorhabditis elegans/drug effects , Cell Line, Tumor , Circular Dichroism , Copper/chemistry , Drug Evaluation, Preclinical , Humans , Iron/chemistry , Iron Chelating Agents , Molecular Dynamics Simulation , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Reactive Oxygen Species/antagonists & inhibitors , Salvia miltiorrhiza , Serum Amyloid A Protein/metabolism , Zinc/chemistry
10.
Mar Drugs ; 11(2): 504-22, 2013 Feb 18.
Article in English | MEDLINE | ID: mdl-23429283

ABSTRACT

We previously reported that a novel marine compound, xyloketal B, has strong antioxidative actions in different models of cardiovascular diseases. Induction of heme oxygenase-1 (HO-1), an important endogenous antioxidant enzyme, has been considered as a potential therapeutic strategy for cardiovascular diseases. We here investigated whether xyloketal B exhibits its antioxidant activity through induction of HO-1. In human umbilical vein endothelial cells (HUVECs), xyloketal B significantly induced HO-1 gene expression and translocation of the nuclear factor-erythroid 2-related factor 2 (Nrf-2) in a concentration- and time-dependent manner. The protection of xyloketal B against angiotensin II-induced apoptosis and reactive oxygen species (ROS) production could be abrogated by the HO-1 specific inhibitor, tin protoporphyrin-IX (SnPP). Consistently, the suppressive effects of xyloketal B on NADPH oxidase activity could be reversed by SnPP in zebrafish embryos. In addition, xyloketal B induced Akt and Erk1/2 phosphorylation in a concentration- and time-dependent manner. Furthermore, PI3K inhibitor LY294002 and Erk1/2 inhibitor U0126 suppressed the induction of HO-1 and translocation of Nrf-2 by xyloketal B, whereas P38 inhibitor SB203580 did not. In conclusion, xyloketal B can induce HO-1 expression via PI3K/Akt/Nrf-2 pathways, and the induction of HO-1 is mainly responsible for the antioxidant and antiapoptotic actions of xyloketal B.


Subject(s)
Endothelial Cells/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Heme Oxygenase-1/metabolism , Pyrans/pharmacology , Zebrafish , Animals , Antioxidant Response Elements , Endothelial Cells/metabolism , Heme Oxygenase-1/genetics , Humans , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Protein Binding , Pyrans/chemistry , Pyrans/metabolism , Respiratory Burst/drug effects , Respiratory Burst/physiology , Reverse Transcriptase Polymerase Chain Reaction
11.
Mar Drugs ; 10(6): 1307-1320, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22822374

ABSTRACT

Cyclotripeptide X-13 is a core of novel marine compound xyloallenoide A isolated from mangrove fungus Xylaria sp. (no. 2508). We found that X-13 dose-dependently induced angiogenesis in zebrafish embryos and in human endothelial cells, which was accompanied by increased phosphorylation of eNOS and Akt and NO release. Inhibition of PI3K/Akt/eNOS by LY294002 or L-NAME suppressed X-13-induced angiogenesis. The present work demonstrates that X-13 promotes angiogenesis via PI3K/Akt/eNOS pathways.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Aquatic Organisms/chemistry , Neovascularization, Physiologic/drug effects , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Angiogenesis Inducing Agents/chemical synthesis , Angiogenesis Inducing Agents/chemistry , Angiogenesis Inducing Agents/isolation & purification , Animals , Biological Products/chemical synthesis , Biological Products/chemistry , Biological Products/isolation & purification , Biological Products/pharmacology , Cell Line , Chromones/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fungi/chemistry , Human Umbilical Vein Endothelial Cells , Humans , Morpholines/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Zebrafish/metabolism
12.
J Cardiovasc Pharmacol ; 59(4): 352-62, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22286127

ABSTRACT

We have previously demonstrated that dl-3n-butylphthalide (NBP) has a potential angiogenic activity. In this study, we investigated the angiogenic effect of NBP and the molecular mechanisms underlying NBP-mediated angiogenesis. Zebrafish embryos and human umbilical vein endothelial cells were treated with various doses of NBP and several signaling pathway inhibitors. NBP induced ectopic subintestinal vessel production in zebrafish embryos and induced invasion, migration, and endothelial cell tube formation of human umbilical vein endothelial cells in a dose-dependent manner. These NBP-induced angiogenic effects were partially suppressed by SU5402, a fibroblast growth factor receptor 1 inhibitor; U0126, an extracellular signal-regulated kinase 1/2 (ERK1/2) inhibitor; LY294002, a phosphatidylinositol 3-kinase inhibitor; 1L6-hydroxymethyl-chiro-inositol-2-(R)-2-O-methyl-3-O-octadecyl-sn-glycerocarbonate, an Akt inhibitor; cavtratin, an endothelial nitric oxide synthase (eNOS) inhibitor and completely inhibited by a combination of U0126 and LY294002. NBP enhanced phosphorylation of ERK1/2 and fibroblast growth factor receptor 2 expression, which were inhibited by U0126. NBP increased the phosphorylation of Akt and eNOS at serine 1177, which was blocked by LY294002. NBP-stimulated nitric oxide production, which was reduced by LY294002. Our data demonstrated that (1) NBP promoted angiogenesis and (2) the angiogenic effects of NBP were mediated by the ERK1/2 and phosphatidylinositol 3-kinase/Akt-eNOS signaling pathways. Our findings suggest that NBP could be a novel agent for therapeutic angiogenesis in ischemic diseases.


Subject(s)
Benzofurans/pharmacology , Neovascularization, Physiologic/drug effects , Neuroprotective Agents/pharmacology , Signal Transduction/drug effects , Animals , Benzofurans/administration & dosage , Dose-Response Relationship, Drug , Human Umbilical Vein Endothelial Cells , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neuroprotective Agents/administration & dosage , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Zebrafish/embryology
13.
Neuroreport ; 22(14): 689-95, 2011 Oct 05.
Article in English | MEDLINE | ID: mdl-21849916

ABSTRACT

Induced pluripotent stem (iPS) cells have been generated from somatic cells by ectopic expression of defined transcription factors. The important issues for clinical applications of iPS cells are the defined methods for somatic cell differentiation and how to effectively enrich desired cell population. Here we used humanized renilla green fluorescent protein under the control of Tα1 α-tubulin promoter as lineage selection marker for neuronal differentiation of iPS cells. Using fluorescence-activated cell sorting, green fluorescent protein positive cells were isolated and enriched to near-purity. These results indicated that the neuronal differentiation potential of iPS cells derived from adult somatic cells is similar to that of embryonic stem cells and the high-purity neurons may have important implications for neurodevelopmental studies, safety pharmacological studies, and transplantation studies.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Age Factors , Animals , Cell Differentiation/physiology , Cells, Cultured , Embryonic Stem Cells/physiology , Induced Pluripotent Stem Cells/physiology , Mice , Mice, Inbred C57BL , Neurons/physiology
14.
Zhong Yao Cai ; 34(8): 1241-6, 2011 Aug.
Article in Chinese | MEDLINE | ID: mdl-22233040

ABSTRACT

OBJECTIVE: To investigate the neuroprotective effects of Lycium barbarum extract against MPP(+) -induced neurotoxicity in Caenorhabditis elegans and PC12 cells and its mechanism. METHODS: Pretreated MPP(+) -induced nearotoxicity in C. elegans and PC12 cells with Lycium barbarum at different dosages. The viability and DA neurodegeneration was assessed in C. elegans selectively expressing green fluorescent protein (GFP) in DA neurons. PC12 cell damage was measured using MTT and nuclear morphology. Intracellular reactive oxygen species (ROS), mitochondrial membrane potential and total GSH were assessed. RESULTS: Lycium barbarum extract protected against MPP(+) -induced loss of viability and DA neurodegeneration in C. elegans in a dose-dependent manner. Similar neuroprotection was replicated in MPP + PC12 cell model. Lycium barbarum extract attenuated MPP(+) -induced intracellular ROS accumulation, loss of mitochondrial membrane potential and restored total GSH levels in PCl2 cells. CONCLUSIONS: Lycium barbarum extract protects against MPP(+) -induced neurotoxicity in C. elegans and PC12 cells and its machanism may be related to its antioxidative property and restoration of total GSH level.


Subject(s)
Lycium/chemistry , Nerve Degeneration/pathology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Plant Extracts/pharmacology , 1-Methyl-4-phenylpyridinium , Animals , Caenorhabditis elegans , Cell Survival/drug effects , Dopamine/metabolism , Dose-Response Relationship, Drug , Flow Cytometry , Glutathione/metabolism , Green Fluorescent Proteins/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/administration & dosage , PC12 Cells , Plant Extracts/administration & dosage , Rats , Reactive Oxygen Species/metabolism
15.
Nan Fang Yi Ke Da Xue Xue Bao ; 30(3): 555-6, 2010 Mar.
Article in Chinese | MEDLINE | ID: mdl-20335136

ABSTRACT

OBJECTIVE: To study the impact of organized stroke ward on the therapeutic effect in stroke patients. METHODS: A total of 2637 patients with acute stroke were randomly assigned to organized stroke ward or the general ward for treatment, and the rates of mortality, nonrecovery, improvement, and recovery were compared between the two groups. RESULTS: The rates of mortality, nonrecovery, improvement, and recovery in 5 years were 2.00%, 0.90%, 74.94% and 22.16% respectively in the organized stroke ward group, as compared to 3.26%, 1.02%, 74.01% and 21.71% in the general ward group, respectively. The mortality rate was significantly lower in organized stroke ward (P<0.05), but no significant difference was found in the rates of nonrecovery, improvement, or recovery between the two groups (P>0.05). CONCLUSION: Admission of the stroke patients in organized stroke ward for treatment can be associated with lowered mortality rate.


Subject(s)
Hospital Units/standards , Intensive Care Units , Stroke/mortality , Stroke/therapy , Female , Humans , Male , Outcome Assessment, Health Care , Patient Care Team/organization & administration , Stroke Rehabilitation , Survival Rate , Treatment Outcome
16.
Brain Res ; 1332: 110-9, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20347725

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disease, affecting 2% of the population over age 65years. Mitochondrial defect and oxidative stress actively participate in the dopaminergic (DA) neuron degeneration in PD. Xyloketal B is a novel marine compound with unique chemical structure isolated from mangrove fungus Xylaria sp. (no. 2508). Recently, we have demonstrated that Xyloketal B can directly scavenge DPPH free radicals and protects mitochondria against oxidative insult. In the present study, we investigate the neuroprotective action of xyloketal B against MPP+-induced neurotoxicity in Caenorhabditis elegans and PC12 cells. The viability and DA neurodegeneration was assessed in C. elegans selectively expressing green fluorescent protein (GFP) in DA neurons. PC12 cell damage was measured using MTT and nuclear morphology. Intracellular reactive oxygen species (ROS), mitochondrial membrane potential and total GSH were assessed. Xyloketal B dose-dependently protected against MPP+-induced loss of viability and DA neurodegeneration in C. elegans. Similar neuroprotection was replicated in MPP+ PC12 cell model. In addition, xyloketal B attenuated MPP+-induced intracellular ROS accumulation, loss of mitochondrial membrane potential and restored total GSH level in PC12 cells. All together, the present study demonstrates that xyloketal B protects against MPP+-induced neurotoxicity in C. elegans and PC12 cells mainly through its antioxidant property and restoration of total GSH level.


Subject(s)
Nerve Degeneration/drug therapy , Neurons/drug effects , Neuroprotective Agents/pharmacology , Pyrans/pharmacology , 1-Methyl-4-phenylpyridinium , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Cell Nucleus/drug effects , Cell Nucleus/pathology , Cell Nucleus/physiology , Cell Survival/drug effects , Dopamine/metabolism , Dose-Response Relationship, Drug , Glutathione/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Membrane Potential, Mitochondrial/drug effects , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Neurons/pathology , Neurons/physiology , Neuroprotective Agents/administration & dosage , PC12 Cells , Pyrans/administration & dosage , Rats , Reactive Oxygen Species/metabolism
17.
Nan Fang Yi Ke Da Xue Xue Bao ; 29(5): 974-8, 2009 May.
Article in Chinese | MEDLINE | ID: mdl-19460724

ABSTRACT

OBJECTIVE: To investigate the differentiation of rat bone marrow mesenchymal stem cells (MSCs) into myocytes and their expression of dystrophin/utrophin after transplantation in mdx mice. METHODS: BrdU-labeled fifth-passage rat MSCs were transplanted in mdx mice with previous total body gamma irradiation (7 Gy). At 4, 8, 12 and 16 weeks after the transplantation, the mice were sacrificed to detect dystrophin/BrdU and utrophin expressions in the gastrocnemius muscle using immunofluorescence assay, RT-PCR and Western blotting. Five normal C57 BL/6 mice and 5 mdx mice served as the positive and negative controls, respectively. RESULTS: Four weeks after MSC transplantation, less than 1% of the muscle fibers of the mdx mice expressed dystrophin, which increased to 15% at 16 weeks. Donor-derived nuclei were detected in both single and clusters of dystrophin-positive fibers. Some BrdU-positive nuclei were centrally located, and some peripherally within myofibers. Utrophin expression decreased over time after transplantation. CONCLUSION: The myofibers of mdx mice with MSC transplantation express dystrophin, which is derived partially from the transplanted MSCs. Dystrophin expression from the transplanted MSCs partially inhibits the upregulation of utrophin in mdx mouse muscle, showing a complementary relation between them.


Subject(s)
Cell Differentiation/physiology , Dystrophin/metabolism , Mesenchymal Stem Cell Transplantation , Muscle Fibers, Skeletal/cytology , Muscular Dystrophy, Animal/therapy , Animals , Bone Marrow Cells/cytology , Dystrophin/genetics , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Inbred mdx/metabolism , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Animal/metabolism , Rats , Utrophin/metabolism
18.
Ann Hematol ; 88(6): 565-72, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19066893

ABSTRACT

Amniotic fluid-derived stem cells have attracted considerable attention in the field of regenerative medicine. Approach of genetic modification probably enhances their regenerative potential. In this work, we wanted to determine whether baculovirus as a new gene vector could efficiently and safely transduce mouse amniotic fluid-derived stem cells (mAFSs). Cells were isolated from mouse amniotic fluid and cultured in vitro. These cells were analyzed by examining phenotypes and differentiation potential. They were further transduced with baculovirus. Baculovirus-transduced mAFSs were induced to differentiate into adipogenic, osteogenic, myogenic, and neurogenic lineages. Mouse amniotic fluid-derived stem cells were successfully isolated and cultured in vitro. They were positive for CD29 and Sca-1, but negative for CD34, CD45, or CD11b. Furthermore, they could differentiate into adipocytes, osteocytes, myocytes, and neurocytes in vitro. Baculovirus could efficiently transduced mAFSs. More importantly, baculovirus-transduced mAFSs retained differentiation potential. Thus, baculovirus vector effective and safe transduction is an attractive promise for genetic modification of mAFSs. Baculovirus genetically modified mAFSs will probably be more suitable as vehicles for regenerative medicine.


Subject(s)
Amniotic Fluid/cytology , Baculoviridae/genetics , Cell Differentiation , Stem Cells/cytology , Stem Cells/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Cell Separation , Cells, Cultured , Mice , Mice, Inbred C57BL , Muscle Development , Neurons/cytology , Osteogenesis , Phenotype
19.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 24(11): 1062-5, 2008 Nov.
Article in Chinese | MEDLINE | ID: mdl-18992192

ABSTRACT

AIM: To study the effects of BMP-2 and FGF-2 on osteoblast differentiation of murine MSCs in vitro. METHODS: The bone marrow cells were collected from 3-18 month old C57BL/6J mice (50 mice), and they were isolated, enriched and expanded using bone marrow adherent culture, and then purified by immunomagnetic microbeads. At last they were identified as mesenchymal stem cells (MSCs). After the MSCs were cultured adherently for 24 hours, 100 microg/L BMP-2 and 0.5 nmol/L FGF-2 were added into osteogenic media separately for 7, 14 and 21 days, alkaline phosphatase (ALP) staining, alkaline phosphatase activities, Vonkossa staining, and Alizarin red S staining were performed step by step. Osteoblast differentiation marker genes including Runx2/cbfa1, ALP, collagen-1, and osteocalcin were investigated by RT real time PCR. RESULTS: ALP activities, calcium deposition and the osteoblast differentiation marker genes of BMP-2 group were markedly higher than those of osteogenic group. The mRNA of Runx2/cbfa1, ALP, collagen-1, and osteocalcin of BMP-2 group was highly expressed than that of osteogenic group. It was the same with FGF-2 group, but less evident than BMP-2 group. CONCLUSION: BMP-2 and FGF-2 can induce murine MSCs to differentiate into osteoblast at different degrees.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Cell Differentiation/drug effects , Fibroblast Growth Factor 2/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Osteoblasts/cytology , Osteogenesis/drug effects , Adaptor Proteins, Signal Transducing/genetics , Animals , Bone Marrow Cells/cytology , Collagen/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Membrane Proteins , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteocalcin/genetics , Polymerase Chain Reaction
20.
Sheng Li Xue Bao ; 60(3): 431-6, 2008 Jun 25.
Article in Chinese | MEDLINE | ID: mdl-18560737

ABSTRACT

The use of stem cells will lead to novel treatments for a wide range of diseases due to their properties of self-renewing, pluripotent, and undifferentiated state, and the stem cells are usually genetically modified for cell and gene therapy. If the baculovirus, as a new gene vector, can be effectively transduced into various mammalian bone marrow-derived mesenchymal stem cells (BMSCs) in vitro, it will be a better gene vector to genetically modify the stem cells. The aim of the present study is to investigate the transduction efficiency of recombinant baculovirus (BacV-CMV-EGFP), which expressed a reporter gene encoding enhanced green fluorescent protein (EGFP) under a cytomegalovirus immediate early (CMV-IE) promoter, into various mammalian BMSCs. The BMSCs of mouse, rat, porcine, rhesus, and human were cultured primarily in vitro. After more than three passages, the mammalian BMSCs were seeded into dishes and cultured in a humidified incubator at 37 °C with 5% CO(2). When the cells reached about 80% confluence, the complete medium was removed by aspiration. The cells were transduced with recombinant baculovirus at a multiplicity of infection (MOI) of 200 vector genomes/cell with 500 µL PBS at 25 °C for 4 h. At the end of baculovirus transduction, cells were washed and incubated with 2 mL complete medium, and baculovirus-transduced mammalian BMSCs were cultured in a humidified incubator for 2 d. Then, the inverted fluorescent microscope was used to observe GFP expressions in different mammalian BMSCs, and flow cytometry was used to detect the transduction efficiency of baculovirus in various mammalian BMSCs. After more than three passages, the BMSCs of mouse, rat, porcine, rhesus, and human showed a homogeneous spindle-shaped morphology. Compared with the BMSCs of mouse, rat and porcine, the inverted fluorescent microscope observations showed that there were more BMSCs expressing GFP and greater mean fluorescence intensity in rhesus and human transduced with baculovirus. The baculovirus could efficiently transduce into the BMSCs of mouse, rat, porcine, rhesus and human, and the transduction efficiency was (20.21±3.02)%, (22.51±4.48)%, (39.13±5.79)%, (71.16±5.36)% and (70.67±3.74)%, respectively. In conclusion, baculovirus displays different transduction efficiency into various mammalian BMSCs. Due to the high transduction efficiency for primate and human BMSCs, baculovirus is possibly a more suitable gene vector to genetically modify BMSCs of human and primates.


Subject(s)
Baculoviridae , Genetic Vectors , Mesenchymal Stem Cells/cytology , Transduction, Genetic , Animals , Bone Marrow Cells/cytology , Genes, Reporter , Green Fluorescent Proteins/genetics , Humans , Macaca mulatta , Mice , Promoter Regions, Genetic , Rats , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...