Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
ACS Omega ; 9(20): 22423-22435, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38799353

ABSTRACT

This study presents the in situ synthesis of CNT@SrTiO3 nanocomposite films for the development of high-performance flexible supercapacitors. The synthesis process involved the use of organic-inorganic hybrid polymers containing metal elements as precursors for thermal decomposition reaction under a reducing atmosphere. Due to the formation of chemical bonding between Ti elements and the CNTs, the interface between STO and CNT surface could provide additional active sites for ion transport and storage. Thereby, the incorporation of SrTiO3 nanoparticles into CNTs enhanced the electrochemical performance of the resulting nanocomposite membranes. To further investigate the influence of STO content and synthesis temperature, we conducted a detailed analysis. The findings indicated that the CNT@STO film with 25% STO content, synthesized at 700 °C, and possessed optimal performance with an areal capacitance of 6682 mF·cm-2 at 5 mV·s-1. Furthermore, a symmetrical flexible supercapacitor assembled by two CNT@STO-25 electrodes demonstrated strong application potential in wearable devices, owing to its long cycle life, excellent flexibility, and high energy density of 430.2 µWh·cm-2 (corresponding power density of 4.5 mW·cm-2). Based on these results, we believe that this study provides a fresh idea for the development of novel flexible energy storage materials.

3.
Clin Cancer Res ; 27(20): 5492-5501, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34253577

ABSTRACT

PURPOSE: Although Bruton tyrosine kinase (BTK) inhibitors have demonstrated promising efficacy in patients with Waldenström macroglobulinemia (WM), data in Asian populations are scarce. This trial is the first to investigate the effect of a BTK inhibitor in Chinese patients with relapsed/refractory (R/R) WM. PATIENTS AND METHODS: Patients with R/R WM with at least one prior regimen were enrolled into this single-arm, multicenter, phase II study (NCT03332173) and received zanubrutinib 160 mg twice daily until disease progression or unacceptable toxicity. The primary endpoint was major response rate (MRR), as assessed by an independent review committee. Secondary endpoints included progression-free survival, overall response rate, duration of major response, and safety. RESULTS: Forty-four patients were enrolled. After a median follow-up of 33.0 (range, 3.2-36.5) months, MRR in all patients was 69.8%, with very good partial response or better in 32.6% of patients. All mutation groups benefited from zanubrutinib treatment (MRR in patients with MYD88 L265P mutation, 73%; MRR in patients with MYD88 wild type mutation, 50%). A higher response rate was seen in the MYD88 L265P/CXCR4 WT population, compared with the other populations. Median progression-free survival and median duration of major response were not reached. The most frequently reported grade ≥3 treatment-emergent adverse events (AEs) were neutrophil count decreased (31.8%), and platelet count decreased and pneumonia (20.5% each). No case of atrial fibrillation/flutter occurred. CONCLUSIONS: Zanubrutinib achieved a high rate of response that was durable and deep in patients with R/R WM across all subgroups, and potentially confers a positive benefit-risk profile for WM.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Piperidines/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Waldenstrom Macroglobulinemia/drug therapy , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Recurrence
4.
BMC Cardiovasc Disord ; 20(1): 339, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32680480

ABSTRACT

BACKGROUND: The efficacy and safety of ambrisentan has been previously evaluated in Chinese patients with pulmonary arterial hypertension (PAH). This post-hoc analysis assessed the efficacy and safety of ambrisentan in a subgroup of connective tissue disease (CTD) patients with PAH. METHODS: In this open-label, single-arm study, patients received ambrisentan 5 mg once daily for 12 weeks, followed by 12-week dose titration period (dose up to 10 mg). Efficacy endpoints included change from baseline in exercise capacity (measured by 6-min walk test [6MWT]), N-terminal pro B type natriuretic peptide (NT-proBNP) plasma levels, WHO Functional Class (FC) and Borg Dyspnoea Index (BDI) scores from baseline to weeks 12 and 24. Safety endpoints included time to clinical worsening and incidence of adverse events (AEs). RESULTS: In total, 71 Chinese patients with CTD-PAH were included in this analysis. Ambrisentan treatment significantly improved exercise capacity (6MWT) from baseline (mean: 366.4 m) to week 12 (63.8 m, p < 0.001) and week 24 (73.2 m, p < 0.001). A significant reduction in NT-proBNP levels was observed from baseline (mean: 1837.5 ng/L) to week 12 (- 1156.8 ng/L, p < 0.001) and week 24 (- 1095.5 ng/L, p < 0.001). BDI scores decreased significantly at week 12 (- 0.6, p < 0.001) and week 24 (- 0.4, p = 0.002) from baseline (mean: 2.7). The WHO FC improved in 29 (40.8%) and 34 (47.9%) patients at weeks 12 and 24, respectively. Adverse events were reported in 52 (73.2%) patients. One patient (1.4%) experienced clinical worsening at week 24. CONCLUSION: Ambrisentan showed significant improvement in exercise capacity and no clinical worsening in the majority of Chinese patients with CTD-PAH in the 24-week treatment period. The AEs observed in the CTD-PAH subgroup were consistent with the known safety profile of ambrisentan in the overall Chinese PAH population. TRIAL REGISTRATION: ClinicalTrial.gov Identifier, https://clinicaltrials.gov/, NCT01808313 Registration date (first time): February 28, 2013.


Subject(s)
Antihypertensive Agents/therapeutic use , Connective Tissue Diseases/complications , Endothelin A Receptor Antagonists/therapeutic use , Phenylpropionates/therapeutic use , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/etiology , Pyridazines/therapeutic use , Adult , Antihypertensive Agents/adverse effects , Beijing , Biomarkers/blood , Connective Tissue Diseases/diagnosis , Endothelin A Receptor Antagonists/adverse effects , Exercise Tolerance/drug effects , Female , Humans , Male , Middle Aged , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Phenylpropionates/adverse effects , Pulmonary Arterial Hypertension/diagnosis , Pulmonary Arterial Hypertension/physiopathology , Pyridazines/adverse effects , Recovery of Function , Time Factors , Treatment Outcome , Young Adult
5.
J Affect Disord ; 257: 143-149, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31301615

ABSTRACT

BACKGROUND: This study evaluated the non-inferiority of bupropion extended-release (XL) compared to escitalopram for acute-phase treatment of Chinese patients with major depressive disorder (MDD). METHODS: This randomized (1:1), double-blind, active-control study conducted between February 2015 and October 2016 included patients with MDD (DSM-IV) (N = 538). The treatment phase had three dose levels (level 1 [Week 1], level 2 [Week 2-4], and level 3 [Week 5-8]), which included either bupropion XL 150 mg, 300 mg, 300 mg or escitalopram 10 mg, 10 mg, 10-20 mg (once-daily), respectively. Primary outcome was mean change from baseline in Hamilton Depression Rating Scale-17 (HAMD-17) total score at Week 8. RESULTS: Overall, 534 patients (bupropion XL, n = 266; escitalopram, n = 268) received at least one dose of study medication. The least square mean (standard error) change from baseline in HAMD-17 total score at Week 8 was -14.5 (0.41) in bupropion XL group and -15.4 (0.39) in escitalopram group (mean difference: 0.8 [-0.27, 1.94]). The response rate was 69.6% versus 72.9%, remission rate was 39.7% versus 47.2%, sustained response rate was 51.6% versus 56.3%, and sustained remission rate was 25.5% versus 28.6%, respectively, for bupropion XL versus escitalopram group. Adverse events were reported by 313 patients (bupropion XL, n = 157; escitalopram, n = 156); the most common on-treatment adverse event in both groups was nausea (10.5% versus 18.7%, respectively). LIMITATIONS: A non-inferiority short-term (8 weeks) study without a placebo arm. CONCLUSION: Results from this study demonstrated that the efficacy of bupropion XL was non-inferior to that of escitalopram in Chinese patients with MDD.


Subject(s)
Antidepressive Agents, Second-Generation/administration & dosage , Bupropion/administration & dosage , Citalopram/administration & dosage , Depressive Disorder, Major/drug therapy , Adult , Asian People/psychology , China , Delayed-Action Preparations , Double-Blind Method , Female , Humans , Least-Squares Analysis , Male , Middle Aged , Treatment Outcome , Young Adult
6.
Eur J Drug Metab Pharmacokinet ; 44(3): 339-352, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30520001

ABSTRACT

BACKGROUND AND OBJECTIVE: Bupropion is used for the treatment of major depressive disorder. We determined the pharmacokinetics, safety, and tolerability of extended-release bupropion XL in healthy Chinese volunteers. METHODS: This open-label, single-center pharmacokinetic study was conducted between May 2016 and June 2016. Eligible volunteers received bupropion XL 150 mg once daily for 5 days, then 300 mg once daily from days 6 to 14. Pharmacokinetic parameters were evaluated after first and repeated doses by non-compartmental and population pharmacokinetic analyses. RESULTS: Fifteen out of 16 enrolled volunteers completed the study. The geometric mean of the bupropion area under the concentration-time curve from 0 to 24 h (AUC0-24) was 498.2 and 1,165.7 h·ng/mL on days 1 and 14, respectively; maximum plasma concentration (Cmax) was 49.9 ng/mL on day 1 and steady-state maximum observed plasma concentration (Css_max) was 111.9 ng/mL on day 14. Among the three metabolites, hydroxybupropion showed the highest AUC0-24 and Cmax. The population pharmacokinetic model findings indicated an apparent oral clearance of 221 L/h for bupropion in a typical healthy 60.9-kg Chinese volunteer. CONCLUSIONS: This was the first pharmacokinetic study for bupropion XL and its active metabolites in the Chinese population. The AUC and Cmax of bupropion XL and its three metabolites increased approximately in a dose-proportional manner with an increase from 150 mg to 300 mg. Adverse events were similar to those reported in studies outside China. A population pharmacokinetic model was developed for bupropion XL, with pharmacokinetics of bupropion adequately described by a two-compartment model with first-order absorption and linear elimination plus lag time. TRIAL REGISTRATION NUMBER: NCT02698553.


Subject(s)
Antidepressive Agents, Second-Generation/adverse effects , Antidepressive Agents, Second-Generation/pharmacokinetics , Bupropion/adverse effects , Bupropion/pharmacokinetics , Models, Biological , Administration, Oral , Adult , Antidepressive Agents, Second-Generation/administration & dosage , Area Under Curve , Asian People , Bupropion/administration & dosage , China , Delayed-Action Preparations , Depressive Disorder, Major/drug therapy , Dose-Response Relationship, Drug , Female , Half-Life , Healthy Volunteers , Humans , Male
7.
Mol Med Rep ; 18(4): 3809-3817, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30132570

ABSTRACT

In contrast with mature brain­derived neurotrophic factor (mBDNF), proBDNF induces cell apoptosis. However, the function of proBDNF in oligodendrocytes remains unclear. In the present study, the OLN­93 oligodendroglia cell line was utilized as an in vitro model to analyse the functions of proBDNF in oligodendroglia. p75NTR, sortilin and proBDNF were expressed in cultured OLN­93 cells. It was indicated that proBDNF inhibited OLN­93 cell proliferation in a dose­dependent manner as determined using the MTT assay and BrdU staining. Furthermore, proBDNF suppressed the migration of OLN­93 cells as demonstrated using the scratch assay. proBDNF also decreased cell viability and promoted apoptosis as indicated by activated cysteine­aspartic acid protease­3 (caspase­3) immunocytochemistry. Notably, anti­proBDNF treatment neutralized the effect of proBDNF and resulted in increased cell proliferation and migration and decreased apoptosis. However, these effects were not observed in the presence of recombinant p75NTR extracellular domain­human FC fusion protein and p75NTR antibody, indicating that proBDNF imparts its inhibitory effects on oligodendrocytes through the p75NTR signal pathway.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cell Movement , Cell Proliferation , Oligodendroglia/cytology , Animals , Apoptosis , Cell Line , Humans , Oligodendroglia/metabolism , Rats , Signal Transduction
8.
J Neurochem ; 144(3): 302-317, 2018 02.
Article in English | MEDLINE | ID: mdl-28869759

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a progressive deposition of amyloid beta (Aß) and dysregulation of neurotrophic signaling, causing synaptic dysfunction, loss of memory, and cell death. The expression of p75 neurotrophin receptor is elevated in the brain of AD patients, suggesting its involvement in this disease. However, the exact mechanism of its action is not yet clear. Here, we show that p75 interacts with beta-site amyloid precursor protein cleaving enzyme-1 (BACE1), and this interaction is enhanced in the presence of Aß. Our results suggest that the colocalization of BACE1 and amyloid precursor protein (APP) is increased in the presence of both Aß and p75 in cortical neurons. In addition, the localization of APP and BACE1 in early endosomes is increased in the presence of Aß and p75. An increased phosphorylation of APP-Thr668 and BACE1-Ser498 by c-Jun N-terminal kinase (JNK) in the presence of Aß and p75 could be responsible for this localization. In conclusion, our study proposes a potential involvement in amyloidogenesis for p75, which may represent a future therapeutic target for AD. Cover Image for this Issue: doi. 10.1111/jnc.14163.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/metabolism , Cerebral Cortex/metabolism , Endosomes/metabolism , Neurons/metabolism , Receptors, Nerve Growth Factor/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Mice, Knockout , Primary Cell Culture , Receptors, Nerve Growth Factor/genetics , Signal Transduction
9.
J Alzheimers Dis ; 59(3): 941-949, 2017.
Article in English | MEDLINE | ID: mdl-28697556

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is pathologically known for the amyloid-ß (Aß) deposition, neurofibrillary tangles, and neuronal loss in the brain. The precursor of brain-derived neurotrophic factor (proBDNF) before proteolysis has opposing functions to its mature form in neuronal survival and neurite growth. However, the role of proBDNF in the pathogenesis of AD remains unclear. OBJECTIVE: To investigate the effects of proBDNF on neurons in vitro, and on learning and memory impairment and brain Aß production in a transgenic AD mouse model (APPswePS1dE9). METHODS: We here examined the effects of proBDNF on the viability (MTT assay) and neurite growth (morphologic measurement) of the primary neurons in vitro. After the intracerebroventricular injection of adeno-associated virus-proBDNF (AAV-proBDNF), we then investigated the learning and memory impairment (Morris water maze) and Aß deposition in the brains of the AD mice. RESULTS: The results showed that proBDNF could inhibit neuronal viability and neurite growth in vitro, enhance Aß levels, and accelerate its deposition in the brain, which was consistent with the learning and memory impairment of AD mice, likely dependent on the membrane receptor of p75NTR. CONCLUSIONS: Our findings suggest that proBDNF may exert a crucially negative effect during AD pathogenesis andprogression.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain/metabolism , Learning Disabilities/metabolism , Memory Disorders/metabolism , Protein Precursors/metabolism , Alzheimer Disease/complications , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Brain-Derived Neurotrophic Factor/therapeutic use , Cells, Cultured , Disease Models, Animal , Female , Hippocampus/cytology , Injections, Intraventricular , Learning Disabilities/etiology , Male , Maze Learning , Memory Disorders/drug therapy , Memory Disorders/etiology , Mice , Mice, Transgenic , Mutation/genetics , Neurons/drug effects , Neurons/metabolism , Presenilin-1/genetics , Protein Precursors/therapeutic use , Receptors, Nerve Growth Factor/deficiency , Receptors, Nerve Growth Factor/genetics , Transduction, Genetic
10.
Proc Natl Acad Sci U S A ; 112(16): 5225-30, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25847999

ABSTRACT

Alzheimer's disease (AD) is one of most devastating diseases affecting elderly people. Amyloid-ß (Aß) accumulation and the downstream pathological events such as oxidative stress play critical roles in pathogenesis of AD. Lessons from failures of current clinical trials suggest that targeting multiple key pathways of the AD pathogenesis is necessary to halt the disease progression. Here we show that Edaravone, a free radical scavenger that is marketed for acute ischemic stroke, has a potent capacity of inhibiting Aß aggregation and attenuating Aß-induced oxidation in vitro. When given before or after the onset of Aß deposition via i.p. injection, Edaravone substantially reduces Aß deposition, alleviates oxidative stress, attenuates the downstream pathologies including Tau hyperphosphorylation, glial activation, neuroinflammation, neuronal loss, synaptic dysfunction, and rescues the behavioral deficits of APPswe/PS1 mice. Oral administration of Edaravone also ameliorates the AD-like pathologies and memory deficits of the mice. These findings suggest that Edaravone holds a promise as a therapeutic agent for AD by targeting multiple key pathways of the disease pathogenesis.


Subject(s)
Alzheimer Disease/drug therapy , Antipyrine/analogs & derivatives , Cognition Disorders/drug therapy , Administration, Oral , Alzheimer Disease/complications , Alzheimer Disease/pathology , Amyloid/metabolism , Amyloid beta-Peptides/toxicity , Animals , Antipyrine/administration & dosage , Antipyrine/chemistry , Antipyrine/pharmacology , Antipyrine/therapeutic use , Behavior, Animal/drug effects , Brain/drug effects , Brain/pathology , Cell Line , Cognition Disorders/complications , Cognition Disorders/pathology , Dendrites/drug effects , Dendrites/pathology , Edaravone , Humans , Inflammation/pathology , Mice, Transgenic , Neurotoxins/toxicity , Oxidative Stress/drug effects , Phosphorylation/drug effects , Presenilin-1/metabolism , Protein Aggregation, Pathological/complications , Protein Aggregation, Pathological/drug therapy , Protein Processing, Post-Translational/drug effects , tau Proteins/metabolism
11.
J Neurochem ; 134(1): 75-85, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25824396

ABSTRACT

Mature brain-derived neurotrophic factor (mBDNF) plays a vital role in the nervous system, whereas proBDNF elicits neurodegeneration and neuronal apoptosis. Although current enzyme-linked immunosorbent assay (ELISA) has been widely used to measure BDNF levels, it cannot differentiate mBDNF from proBDNF. As the function of proBDNF differs from mBDNF, it is necessary to establish an ELISA assay specific for the detection of mBDNF. Therefore, we aimed to establish a new mBDNF-specific sandwich ELISA. In this study, we have screened and found a combination of antibodies for a sandwich ELISA. A monoclonal antibody and sheep anti-BDNF were chosen as capture and detection antibody for sandwich ELISA respectively. The new ELISA showed no cross-reactivity to human recombinant NT-3, NT-4, nerve growth factor and negligible cross-reactivity (0.99-4.99%) for proBDNF compared to commercial ELISA kits (33.18-91.09%). The application of the new mBDNF ELISA was shown through the measurement of mBDNF levels in different brain regions of rats and in the brain of ß-site amyloid precursor protein cleaving enzyme 1 (BACE1)(-/-) and WT mice and compared to western blot. Overall, this new ELISA will be useful for the measurement of mBDNF levels with high specificity. As the function of proBDNF differs from mBDNF (mature BDNF), it is necessary to establish an ELISA specific for the detection of mBDNF. Here, we present a novel sandwich ELISA which detects mBDNF with high specificity. This new ELISA will be useful for the measurement of mBDNF levels with high specificity in various human and animal tissues. proBDNF, precursor of BDNF; BDNF, brain-derived neurotrophic factor; NT-3, neurotrophin-3; NT-4, neurotrophin-4; NGF, nerve growth factor.


Subject(s)
Brain-Derived Neurotrophic Factor/analysis , Brain-Derived Neurotrophic Factor/biosynthesis , Enzyme-Linked Immunosorbent Assay/trends , Animals , Brain-Derived Neurotrophic Factor/genetics , Enzyme-Linked Immunosorbent Assay/standards , Female , Humans , Male , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Sheep
12.
J Neurochem ; 127(2): 152-62, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23895422

ABSTRACT

Sortilin, a Golgi sorting protein and a member of the VPS10P family, is the co-receptor for proneurotrophins, regulates protein trafficking, targets proteins to lysosomes, and regulates low density lipoprotein metabolism. The aim of this study was to investigate the expression and regulation of sortilin in Alzheimer's disease (AD). A significantly increased level of sortilin was found in human AD brain and in the brains of 6-month-old swedish-amyloid precursor protein/PS1dE9 transgenic mice. Aß42 enhanced the protein and mRNA expression levels of sortilin in a dose- and time-dependent manner in SH-SY5Y cells, but had no effect on sorLA. In addition, proBDNF also significantly increased the protein and mRNA expression of sortilin in these cells. The recombinant extracellular domain of p75(NTR) (P75ECD-FC), or the antibody against the extracellular domain of p75(NTR), blocked the up-regulation of sortilin induced by Amyloid-ß protein (Aß), suggesting that Aß42 increased the expression level of sortilin and mRNA in SH-SY5Y via the p75(NTR) receptor. Inhibition of ROCK, but not Jun N-terminal kinase, suppressed constitutive and Aß42-induced expression of sortilin. In conclusion, this study shows that sortilin expression is increased in the AD brain in human and mice and that Aß42 oligomer increases sortilin gene and protein expression through p75(NTR) and RhoA signaling pathways, suggesting a potential physiological interaction of Aß42 and sortilin in Alzheimer's disease.


Subject(s)
Adaptor Proteins, Vesicular Transport/biosynthesis , Amyloid beta-Peptides/physiology , Peptide Fragments/physiology , Receptor, Nerve Growth Factor/biosynthesis , rhoA GTP-Binding Protein/metabolism , Aged, 80 and over , Alzheimer Disease/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/biosynthesis , Animals , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Humans , Mice , Mice, Transgenic , Peptide Fragments/genetics , Presenilin-1/biosynthesis , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Up-Regulation/physiology
13.
Biometrics ; 69(3): 758-65, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23865502

ABSTRACT

Estimating population sizes via capture-recapture experiments has enormous applications. The Poisson mixture model can be adopted for those applications with a single list in which individuals appear one or more times. We compare several nonparametric estimators, including the Chao estimator, the Zelterman estimator, two jackknife estimators and the bootstrap estimator. The target parameter of the Chao estimator is a lower bound of the population size. Those of the other four estimators are not lower bounds, and they may produce lower confidence limits for the population size with poor coverage probabilities. A simulation study is reported and two examples are investigated.


Subject(s)
Models, Statistical , Poisson Distribution , Population Density , Bias , Biometry/methods , Computer Simulation , Confidence Intervals , Heroin Dependence/epidemiology , Humans , Likelihood Functions , Statistics, Nonparametric , Thailand/epidemiology , Western Australia/epidemiology
14.
PLoS One ; 8(5): e63049, 2013.
Article in English | MEDLINE | ID: mdl-23704887

ABSTRACT

The processing of Amyloid precursor protein (APP) is multifaceted, comprising of protein transport, internalization and sequential proteolysis. However, the exact mechanism of APP intracellular trafficking and distribution remains unclear. To determine the interaction between sortilin and APP and the effect of sortilin on APP trafficking and processing, we studied the binding site and its function by mapping experiments, colocalization, coimmunoprecipitation and sucrose gradient fractionation. We identified for the first time that sortilin interacts with APP at both N- and C-terminal regions. The sortilin-FLVHRY (residues 787-792) and APP-NPTYKFFE (residues 759-766) motifs are crucial for the C-terminal interaction. We also found that lack of the FLVHRY motif reduces APP lysosomal targeting and increases APP distribution in lipid rafts in co-transfected HEK293 cells. These results are consistent with our in vivo data where sortilin knockout mice showed a decrease of APP lysosomal distribution and an increase of APP in lipid rafts. We further confirmed that overexpression of sortilin-FLVHRY mutants failed to rescue the lysosomal degradation of APP. Thus, our data suggests that sortilin is implicated in APP lysosomal and lipid raft targeting via its carboxyl-terminal F/YXXXXF/Y motif. Our study provides new molecular insights into APP trafficking and processing.


Subject(s)
Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/metabolism , Amyloid beta-Protein Precursor/metabolism , Lysosomes/metabolism , Membrane Microdomains/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cerebral Cortex/cytology , Endosomes/metabolism , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Mice , Molecular Sequence Data , Neurons/metabolism , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary , Protein Transport , Proteolysis , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Structure-Activity Relationship , Transfection
15.
Neuro Oncol ; 15(8): 990-1007, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23576602

ABSTRACT

BACKGROUND: High-grade glioma is incurable, with a short survival time and poor prognosis. The increased expression of p75 neurotrophin receptor (NTR) is a characteristic of high-grade glioma, but the potential significance of increased p75NTR in this tumor is not fully understood. Since p75NTR is the receptor for the precursor of brain-derived neurotrophic factor (proBDNF), it is suggested that proBDNF may have an impact on glioma. METHODS: In this study we investigated the expression of proBDNF and its receptors p75NTR and sortilin in 52 cases of human glioma and 13 cases of controls by immunochemistry, quantitative real-time PCR, and Western blot methods. Using C6 glioma cells as a model, we investigated the roles of proBDNF on C6 glioma cell differentiation, growth, apoptosis, and migration in vitro. RESULTS: We found that the expression levels of proBDNF, p75NTR, and sortilin were significantly increased in high-grade glioma and were positively correlated with the malignancy of the tumor. We also observed that tumors expressed proBDNF, p75NTR, and sortilin in the same cells with different subcellular distributions, suggesting an autocrine or paracrine loop. The ratio of proBDNF to mature BDNF was decreased in high-grade glioma tissues and was negatively correlated with tumor grade. Using C6 glioma cells as a model, we found that proBDNF increased apoptosis and differentiation and decreased cell growth and migration in vitro via p75NTR. CONCLUSIONS: Our data indicate that proBDNF and its receptors are upregulated in high-grade glioma and might play an inhibitory effect on glioma.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , Brain-Derived Neurotrophic Factor/metabolism , Brain/metabolism , Glioma/pathology , Protein Precursors/metabolism , Receptor, Nerve Growth Factor/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Adult , Apoptosis , Biomarkers, Tumor/genetics , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain-Derived Neurotrophic Factor/genetics , Case-Control Studies , Cell Proliferation , Female , Follow-Up Studies , Glioma/genetics , Glioma/metabolism , Humans , Immunoenzyme Techniques , In Vitro Techniques , Male , Middle Aged , Neoplasm Grading , Prognosis , Protein Precursors/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, Nerve Growth Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
16.
J Affect Disord ; 150(3): 776-84, 2013 Sep 25.
Article in English | MEDLINE | ID: mdl-23537780

ABSTRACT

BACKGROUND: In recent decades, the role of brain-derived neurotrophic factor (BDNF) in depression has received intensive attention. However, the relationship between proBDNF and depression has not been clearly elucidated. METHODS: Forty drug-free women patients diagnosed with major depression and 50 healthy female controls were enrolled in our study. Peripheral blood was sampled from all the subjects. With the blood samples, we assessed the relationship between BDNF and major depression from following aspects: the levels of BDNF, proBDNF and their receptors in the sera and lymphocytes. The mRNA levels of these factors in lymphocytes were also examined. Furthermore, the correlations between each factor and the severity of major depression were tested. RESULTS: It was found that: (a) the protein and serum levels of proBDNF, sortilin and p75NTR were higher in major depressive patients than in healthy controls while mature BDNF and TrkB levels were lower; (b) the BDNF, TrkB, sortilin and p75NTR mRNA levels changed in line with their protein levels; (c) The levels of mature BDNF and TrkB had negative correlations with the major depression severity, and the levels of proBDNF, p75NTR and sortilin were positively correlated with the scores of HRSD-21; (d) the ratio of proBDNF and mBDNF was imbalanced in major depressive patients. CONCLUSION: The balance between the proBDNF/p75NTR/sortilin and mBDNF/TrkB signaling pathways appears dysregulated in major depression and both pathways should be considered as biomarkers for the major depression LIMITATIONS: More cases on both genders should be enrolled in our study. And further works on the mechanisms of how BDNF and its receptors are regulated in depression should also be carried out.


Subject(s)
Brain-Derived Neurotrophic Factor/blood , Depressive Disorder, Major/blood , Protein Precursors/blood , Receptor, trkB/blood , Adaptor Proteins, Vesicular Transport/blood , Adult , Brain-Derived Neurotrophic Factor/biosynthesis , China , Depressive Disorder, Major/metabolism , Female , Humans , Middle Aged , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/blood , Protein Precursors/biosynthesis , RNA, Messenger/biosynthesis , Receptor, trkB/biosynthesis , Receptors, Nerve Growth Factor/biosynthesis , Receptors, Nerve Growth Factor/blood , Signal Transduction , Up-Regulation , Young Adult
17.
J Neurochem ; 122(5): 1010-22, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22731248

ABSTRACT

Amyloid precursor protein (APP) is involved in the pathogenesis of Alzheimer's disease. It is axonally transported, endocytosed and sorted to different cellular compartments where amyloid beta (Aß) is produced. However, the mechanism of APP trafficking remains unclear. We present evidence that huntingtin associated protein 1 (HAP1) may reduce Aß production by regulating APP trafficking to the non-amyloidogenic pathway. HAP1 and APP are highly colocalized in a number of brain regions, with similar distribution patterns in both mouse and human brains. They are associated with each other, the interacting site is the 371-599 of HAP1. APP is more retained in cis-Golgi, trans-Golgi complex, early endosome and ER-Golgi intermediate compartment in HAP1-/- neurons. HAP1 deletion significantly alters APP endocytosis and reduces the re-insertion of APP into the cytoplasmic membrane. Amyloid precursor protein-YFP(APP-YFP) vesicles in HAP1-/- neurons reveal a decreased trafficking rate and an increased number of motionless vesicles. Knock-down of HAP1 protein in cultured cortical neurons of Alzheimer's disease mouse model increases Aß levels. Our data suggest that HAP1 regulates APP subcellular trafficking to the non-amyloidogenic pathway and may negatively regulate Aß production in neurons.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Autoantigens/metabolism , Biotinylation , Brain/metabolism , Cells, Cultured , Cerebral Cortex/pathology , Cytoplasm/metabolism , Disease Models, Animal , Endocytosis/genetics , Endoplasmic Reticulum Chaperone BiP , Enzyme-Linked Immunosorbent Assay , Fluorescence Resonance Energy Transfer/methods , Heat-Shock Proteins/metabolism , Humans , Immunoprecipitation , Integrins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Lysosomal-Associated Membrane Protein 1/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/deficiency , Neurons/ultrastructure , Photobleaching , Presenilin-1/genetics , Presenilin-1/metabolism , Protein Transport/genetics , RNA Interference/physiology , Transfection/methods , Vesicular Transport Proteins/metabolism , trans-Golgi Network/genetics , trans-Golgi Network/metabolism
18.
PLoS One ; 7(4): e35883, 2012.
Article in English | MEDLINE | ID: mdl-22558255

ABSTRACT

BACKGROUND: Neurons extend their dendrites and axons to build functional neural circuits, which are regulated by both positive and negative signals during development. Brain-derived neurotrophic factor (BDNF) is a positive regulator for neurite outgrowth and neuronal survival but the functions of its precursor (proBDNF) are less characterized. METHODOLOGY/PRINCIPAL FINDINGS: Here we show that proBDNF collapses neurite outgrowth in murine dorsal root ganglion (DRG) neurons and cortical neurons by activating RhoA via the p75 neurotrophin receptor (p75NTR). We demonstrated that the receptor proteins for proBDNF, p75NTR and sortilin, were highly expressed in cultured DRG or cortical neurons. ProBDNF caused a dramatic neurite collapse in a dose-dependent manner and this effect was about 500 fold more potent than myelin-associated glycoprotein. Neutralization of endogenous proBDNF by using antibodies enhanced neurite outgrowth in vitro and in vivo, but this effect was lost in p75NTR(-/-) mice. The neurite outgrowth of cortical neurons from p75NTR deficient (p75NTR(-/-)) mice was insensitive to proBDNF. There was a time-dependent reduction of length and number of filopodia in response to proBDNF which was accompanied with a polarized RhoA activation in growth cones. Moreover, proBDNF treatment of cortical neurons resulted in a time-dependent activation of RhoA but not Cdc42 and the effect was absent in p75NTR(-/-) neurons. Rho kinase (ROCK) and the collapsin response mediator protein-2 (CRMP-2) were also involved in the proBDNF action. CONCLUSIONS: proBDNF has an opposing role in neurite outgrowth to that of mature BDNF. Our observations suggest that proBDNF collapses neurites outgrowth and filopodial growth cones by activating RhoA through the p75NTR signaling pathway.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Ganglia, Spinal/drug effects , Gene Expression Regulation, Developmental/physiology , Protein Precursors/physiology , rho GTP-Binding Proteins/agonists , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Antibodies/pharmacology , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Brain-Derived Neurotrophic Factor/pharmacology , Cells, Cultured , Ganglia, Spinal/cytology , Ganglia, Spinal/physiology , Mice , Mice, Knockout , Nerve Fibers/drug effects , Nerve Fibers/physiology , Neurites/drug effects , Neurites/physiology , Protein Precursors/pharmacology , Pseudopodia/drug effects , Pseudopodia/physiology , Receptors, Nerve Growth Factor/deficiency , Receptors, Nerve Growth Factor/genetics , Signal Transduction/physiology , Time-Lapse Imaging , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
19.
J Biol Chem ; 286(18): 16272-84, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21357693

ABSTRACT

proBDNF, a precursor of brain-derived neurotrophic factor (BDNF), is anterogradely transported and released from nerve terminals, but the mechanism underlying this process remains unclear. In this study, we report that proBDNF forms a complex with Huntingtin associated protein-1 (HAP1) and sortilin, which plays an important role in proBDNF intracellular trafficking and stabilization. The interaction of proBDNF with both HAP1A and sortilin in co-transfected HEK293 cells is confirmed by both fluorescence resonance energy transfer and co-immunoprecipitation. The frequent co-localization (>90%) of endogenous HAP1, sortilin, and proBDNF is also found in cultured cortical neurons. Mapping studies using GST pulldown and competition assays has defined the interacting region of HAP1 with proBDNF within amino acids 371-445 and the binding sequences of proBDNF to HAP1 between amino acids 65 and 90. Fluorescence recovery after photobleaching confirms the defective movement of proBDNF-containing vesicles in neurites of HAP1(-/-) neurons, which can be partially restored by reintroducing HAP1 cDNA into the neurons. However, the effect is significantly increased by simultaneously reintroducing both HAP1 and sortilin. proBDNF and HAP1 are highly co-localized with organelle markers for the Golgi network, microtubules, molecular motor, or endosomes in normal neurons, but this co-localization is reduced in HAP1(-/-) neurons. Co-immunoprecipitation and Western blot showed that sortilin stabilizes the proBDNF·HAP1 complex in co-transfected HEK293 cells, helping to prevent proBDNF degradation. Furthermore, the complex facilitates furin cleavage to release mature BDNF.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Nerve Tissue Proteins/metabolism , Protein Precursors/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Animals , Brain-Derived Neurotrophic Factor/genetics , Cerebral Cortex/metabolism , HEK293 Cells , Humans , Mice , Mice, Knockout , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Nerve Tissue Proteins/genetics , Neurons/metabolism , Organelles/genetics , Organelles/metabolism , PC12 Cells , Peptide Mapping/methods , Protein Precursors/genetics , Protein Stability , Protein Transport/physiology , Rats
20.
Eur J Neurosci ; 33(8): 1376-84, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21366730

ABSTRACT

The majority of newborn neurons migrate from their birthplace to final destination in the developing brain. Migration of cerebellar granule cells (CGCs) requires multiple factors. Mature brain-derived neurotrophic factor (BDNF) positively regulates the proliferation, migration, survival and differentiation of CGCs in rodents. However, the role of the BDNF precursor, proBDNF, in neuronal development remains unknown. In this study, we investigated the effect of proBDNF in vivo and in vitro on migration of CGCs. We demonstrate that proBDNF and its receptors p75 neurotrophin receptor (p75NTR) and sortilin are highly expressed in the cerebella as determined by immunohistochemistry and Western blot. ProBDNF is released from cultured cerebellar neurons, and this release is increased by high potassium stimulation. ProBDNF inhibits migration of CGCs in vitro, and the neutralizing antibodies to proBDNF enhance such migration as assayed by transwell culture. In addition, proBDNF incorporated into an agarose plug reduces granule cell migration from such plugs, whereas the neutralizing antibodies attract these cells towards the plug. The application of proBDNF into the lateral ventricle significantly inhibits migration of CGCs out of the proliferative zone into the internal granular cell layer, whereas the neutralizing antibodies enhance this migration. Furthermore, the effects of proBDNF on cell migration are lost in p75NTR(-/-) mice. Our data suggest that proBDNF negatively regulates migration of CGCs and this effect is mediated by p75NTR. We conclude that proBDNF has an opposing role in migration of CGCs to that of mature BDNF.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cell Movement/physiology , Cerebellum/cytology , Neurons/physiology , Protein Precursors/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Cell Movement/drug effects , Cells, Cultured , Cerebellum/drug effects , Cerebellum/metabolism , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/drug effects , Protein Precursors/pharmacology , Receptor, Nerve Growth Factor/genetics , Receptor, Nerve Growth Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...