Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Drugs R D ; 2024 May 30.
Article in English | MEDLINE | ID: mdl-38811485

ABSTRACT

BACKGROUND AND OBJECTIVE: Azithromycin is the first azalide antibiotic that is related to the macrolide family of antibiotics. Bioequivalence studies in China are initiated by the National Medical Products Administration (NMPA), which supports a generic consistency evaluation program for ensuring that generic products manufactured in China meet the required standards and provide equivalent therapeutic effects to their reference products. This study aimed to assess the bioequivalence of two azithromycin tablets under both fasting and fed conditions in healthy Chinese volunteers. METHODS: This was a single-center, open-label, single-dose, randomized, three-way crossover trial with two independent groups (fasting group and fed group). A total of 72 healthy Chinese subjects (36 subjects in the fasting state and 36 subjects in the fed state) were enrolled and randomized to treatment. Blood samples were collected from 0 to 120 h after a single oral dose of a 250-mg generic azithromycin tablet (test, T) or branded azithromycin tablet (reference, R). The plasma concentrations of azithromycin were determined by high-performance liquid chromatography-tandem mass spectrometry (HPLC‒MS/MS). A non-compartmental analysis method was used to estimate the pharmacokinetic parameters. Adverse events were documented. RESULTS: In a fasting state, the bioequivalence of maximum plasma concentration (Cmax) was evaluated using the reference-scaled average bioequivalence (RSABE) approach (within-subject standard deviation, SWR > 0.294), and the bioequivalence of area under the concentration-time curve from time 0 to the time of the last measurable plasma concentration (AUC0-t) and area under the concentration-time curve from time 0 extrapolated to infinity (AUC0-∞) were evaluated by the average bioequivalence (ABE) method (SWR <  0.294). The geometric mean ratio (GMR) of T/R for Cmax was 106.49%, while the 95% upper confidence bound was <  0. The GMRs of AUC0-t and AUC0-∞ were 103.34% and 101.28%, and the 90% confidence intervals (CIs) of the test/reference were 95.90-111.35%/94.85-108.15%, respectively. In the fed state, the RSABE approach was applied to estimate the bioequivalence of Cmax (SWR >0.294), and the ABE approach was applied to estimate the bioequivalence of AUC0-t and AUC0-∞ (SWR <  0.294). The GMR for Cmax was 99.80%, while the 95% upper confidence bound value was <  0. The GMRs of AUC0-t and AUC0-∞ were 97.07% and 98.15%, and the 90% CIs of the T/R were 90.02-104.68% and 90.66-106.25%, respectively. All adverse events were mild and transient. CONCLUSIONS: The trial indicated that the test and the reference azithromycin tablets were bioequivalent and well tolerated in healthy Chinese volunteers under both fasting and fed conditions. TRIAL REGISTRATION: Clinicaltrials, ChiCTR2300071630 (retrospectively registered in 19/05/2023).

2.
Xenobiotica ; 54(1): 38-44, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38085693

ABSTRACT

Gefitinib is the first-generation drug of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) metabolised by the cytochrome P450 and transported by P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2). In the present study, the pharmacokinetics of gefitinib in healthy Chinese volunteers was investigated and the effect of genetic polymorphisms on its variability was evaluted.Forty-five healthy volunteers were administered a single dose of gefitinib and the blood samples were used for quantifying the concentration of gefitinib and genotyping fifteen single-nucleotide polymorphisms of cytochrome P450 enzymes (CYP3A4, CYP3A5, CYP2D6, CYP2C9 and CYP2C19) and drug transporters (ABCB1 and ABCG2).CYP3A5*3 (rs776746) polymorphism showed a significant influence, with higher gefitinib AUC0-t in carrier of CC genotype than in CT/TT genotype (BH-adjusted p value <0.05). For CYP2C9*3 (rs1057910), significant differences in pharmacokinetics of gefitinib were detected between carriers of AA and AC genotypes, with higher AUC0-t, AUC0-∞ and Cmax in carrier of AC genotype than in AA gen-otype (BH-adjusted p value <0.05). No associations were found between SNPs in CYP3A4, CYP2D6, CYP2C19, ABCB1, ABCG2 and the pharmacokinetics of gefitinib.The SNPs in CYP3A5*3 (rs776746) and CYP2C9*3 (rs1057910) were found to be associated with altered gefitinib pharmacokinetics in healthy Chinese volunteers.


Subject(s)
Cytochrome P-450 CYP2D6 , Cytochrome P-450 CYP3A , Humans , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Gefitinib , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2C19/metabolism , Cytochrome P-450 CYP2D6/metabolism , Healthy Volunteers , Cytochrome P-450 CYP2C9/genetics , Cytochrome P-450 CYP2C9/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Polymorphism, Single Nucleotide , Genotype , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , China
3.
Clin Pharmacol Drug Dev ; 11(9): 1084-1091, 2022 09.
Article in English | MEDLINE | ID: mdl-35396924

ABSTRACT

To evaluate the pharmacokinetic properties and bioequivalence of 2 oral formulations of domperidone in healthy Chinese subjects, a randomized, open-label, 2-way crossover study was conducted under fasting and fed states. All 96 healthy subjects were randomized to receive a single oral dose of a 10-mg generic domperidone tablet (test) or branded domperidone tablet (reference). Blood samples were collected at specified time intervals and analyzed for domperidone using liquid chromatography-tandem mass spectrometry. In the fasting test, 90% CIs of geometric mean ratios were 86.7% to 105.8% for maximum concentration, 96.7% to 106.1% for area under the concentration-time curve (AUC) from time 0 to the time of the last measurable plasma concentration, and 97.1% to 106.1% for AUC from time 0 extrapolated to infinity. In the fed test, the 90% CIs were 90.8% to 121.1%, 99.7% to 109.4%, and 99.4% to 109.1%, respectively. All 90% CIs were within the bioequivalence range of 80% to 125%, indicating that the 2 formulations were bioequivalent. In addition, the values of time to maximum concentration, terminal-phase elimination half-life, and AUC were significantly higher in the fed group than in the fasting group, suggesting that a high-fat meal slowed down the absorption and elimination of domperidone and significantly increased domperidone exposure.


Subject(s)
Domperidone , Tandem Mass Spectrometry , Area Under Curve , China , Cross-Over Studies , Healthy Volunteers , Humans , Tablets , Tandem Mass Spectrometry/methods , Therapeutic Equivalency
5.
J Cell Mol Med ; 25(10): 4776-4785, 2021 05.
Article in English | MEDLINE | ID: mdl-33788387

ABSTRACT

As a serious metabolic disease, diabetes causes series of complications that seriously endanger human health. The liver is a key organ for metabolizing glucose and lipids, which substantially contributes to the development of insulin resistance and type 2 diabetes mellitus (T2DM). Exogenous fibroblast growth factor 1 (FGF1) has a great potential for the treatment of diabetes. Receptor of advanced glycation end products (RAGE) is a receptor for advanced glycation end products that involved in the development of diabetes-triggered complications. Previous study has demonstrated that FGF1 significantly ameliorates diabetes-mediated liver damage (DMLD). However, whether RAGE is involved in this process is still unknown. In this study, we intraperitoneally injected db/db mice with 0.5 mg/kg FGF1. We confirmed that FGF1 treatment not only significantly ameliorates diabetes-induced elevated apoptosis in the liver, but also attenuates diabetes-induced inflammation, then contributes to ameliorate liver dysfunction. Moreover, we found that diabetes triggers the elevated RAGE in hepatocytes, and FGF1 treatment blocks it, suggesting that RAGE may be a key target during FGF1 treatment of diabetes-induced liver injury. Thus, we further confirmed the role of RAGE in FGF1 treatment of AML12 cells under high glucose condition. We found that D-ribose, a RAGE agonist, reverses the protective role of FGF1 in AML12 cells. These findings suggest that FGF1 ameliorates diabetes-induced hepatocyte apoptosis and elevated inflammation via suppressing RAGE pathway. These results suggest that RAGE may be a potential therapeutic target for the treatment of DMLD.


Subject(s)
Acute Lung Injury/drug therapy , Diabetes Complications/drug therapy , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/complications , Fibroblast Growth Factor 1/pharmacology , Inflammation/drug therapy , Receptor for Advanced Glycation End Products/antagonists & inhibitors , Acute Lung Injury/etiology , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Apoptosis , Diabetes Complications/etiology , Diabetes Complications/metabolism , Diabetes Complications/pathology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL
6.
Cell Death Dis ; 12(1): 107, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33479232

ABSTRACT

Prolonged type 2 diabetes mellitus (T2DM) produces a common complication, peripheral neuropathy, which is accompanied by nerve fiber disorder, axon atrophy, and demyelination. Growing evidence has characterized the beneficial effects of acidic fibroblast growth factor (aFGF) and shown that it relieves hyperglycemia, increases insulin sensitivity, and ameliorates neuropathic impairment. However, there is scarce evidence on the role of aFGF on remodeling of aberrant myelin under hyperglycemia condition. Presently, we observed that the expression of aFGF was rapidly decreased in a db/db T2DM mouse model. Administration of exogenous aFGF was sufficient to block acute demyelination and nerve fiber disorganization. Furthermore, this strong anti-demyelinating effect was most likely dominated by an aFGF-mediated increase of Schwann cell (SC) proliferation and migration as well as suppression of its apoptosis. Mechanistically, the beneficial biological effects of aFGF on SC behavior and abnormal myelin morphology were likely due to the inhibition of hyperglycemia-induced oxidative stress activation, which was most likely activated by kelch-like ECH-associated protein 1 (Keap1)/nuclear factor erythroid-derived-like 2 (Nrf2) signaling. Thus, this evidence indicates that aFGF is a promising protective agent for relieving myelin pathology through countering oxidative stress signaling cascades under diabetic conditions.


Subject(s)
Diabetes Mellitus, Type 2/therapy , Fibroblast Growth Factor 1/therapeutic use , Animals , Disease Models, Animal , Fibroblast Growth Factor 1/pharmacology , Male , Mice , Oxidative Stress , Rats , Reactive Oxygen Species
7.
Acta Pharmacol Sin ; 42(3): 347-360, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33462377

ABSTRACT

DL-3-n-Butylphthalide (DL-NBP), a small molecular compound extracted from the seeds of Apium graveolens Linn (Chinese celery), has been shown to exert neuroprotective effects due to its anti-inflammatory, anti-oxidative and anti-apoptotic activities. DL-NBP not only protects against ischemic cerebral injury, but also ameliorates vascular cognitive impairment in dementia patients including AD and PD. In the current study, we investigated whether and how DL-NBP exerted a neuroprotective effect against diabetes-associated cognitive decline (DACD) in db/db mice, a model of type-2 diabetes. db/db mice were orally administered DL-NBP (20, 60, 120 mg· kg-1· d-1) for 8 weeks. Then the mice were subjected to behavioral test, their brain tissue was collected for morphological and biochemical analyses. We showed that oral administration of DL-NBP significantly ameliorated the cognitive decline with improved learning and memory function in Morris water maze testing. Furthermore, DL-NBP administration attenuated diabetes-induced morphological alterations and increased neuronal survival and restored the levels of synaptic protein PSD95, synaptophysin and synapsin-1 as well as dendritic density in the hippocampus, especially at a dose of 60 mg/kg. Moreover, we revealed that DL-NBP administration suppressed oxidative stress by upregulating Nrf2/HO-1 signaling, and increased brain-derived neurotrophic factor (BDNF) expression by activating PI3K/Akt/CREB signaling in the hippocampus. These beneficial effects of DL-NBP were observed in high glucose-treated PC12 cells. Our results suggest that DL-NBP may be a potential pharmacologic agent for the treatment of DACD.


Subject(s)
Benzofurans/therapeutic use , Cognitive Dysfunction/drug therapy , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Cognitive Dysfunction/etiology , Dendrites/drug effects , Diabetes Mellitus, Type 2/complications , Hippocampus/drug effects , Male , Mice, Inbred C57BL , Morris Water Maze Test/drug effects , PC12 Cells , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Synapses/drug effects
9.
Cell Commun Signal ; 18(1): 81, 2020 05 27.
Article in English | MEDLINE | ID: mdl-32460803

ABSTRACT

BACKGROUND: Diabetes induces central nervous system damage, leading to cognitive decline. Fibroblast growth factor 1 (FGF1) has dual function of neuroprotection and normalizing hyperglycemia. To date, the precise mechanisms and potential treating strategies of FGF1 for diabetes-induced cognitive decline (DICD) hasn't been fully elucidated. METHODS: In this study, db/db mice were used as DICD animal model. We found that diabetes remarkably suppressed FGF1 expression in hippocampus. Thus, exogenous FGF1 had been treated for db/db mice and SH-SY5Y cells. RESULTS: FGF1 significantly ameliorates DICD with better spatial learning and memory function. Moreover, FGF1 blocked diabetes-induced morphological structure change, neuronal apoptosis and Aß1-42 deposition and synaptic dysfunction in hippocampus. But normalizing glucose may not the only contributed factor for FGF1 treating DICD with evidencing that metformin-treated db/db mice has a inferior cognitive function than that in FGF1 group. Current mechanistic study had found that diabetes inhibits cAMP-response element binding protein (CREB) activity and subsequently suppresses brain derived neurotrophic factor (BDNF) level via coordinately regulating PERK signaling and PI3K/AKT signaling in hippocampus, which were reversed by FGF1. CONCLUSION: We conclude that FGF1 exerts its neuroprotective role and normalizing hyperglycemia effect, consequently ameliorates DICD, implying FGF1 holds a great promise to develop a new treatment for DICD. Video abstract.


Subject(s)
Cognitive Dysfunction/drug therapy , Diabetes Complications/drug therapy , Fibroblast Growth Factor 1/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , eIF-2 Kinase/metabolism , Animals , Cell Line , Cognitive Dysfunction/metabolism , Diabetes Complications/metabolism , Diabetes Mellitus/physiopathology , Humans , Male , Mice , Mice, Inbred C57BL , Signal Transduction
10.
Theranostics ; 10(4): 1649-1677, 2020.
Article in English | MEDLINE | ID: mdl-32042328

ABSTRACT

Rationale: Autophagy in Schwann cells (SCs) is crucial for myelin debris degradation and clearance following peripheral nerve injury (PNI). Nerve growth factor (NGF) plays an important role in reconstructing peripheral nerve fibers and promoting axonal regeneration. However, it remains unclear if NGF effect in enhancing nerve regeneration is mediated through autophagic clearance of myelin debris in SCs. Methods: In vivo, free NGF solution plus with/without pharmacological inhibitors were administered to a rat sciatic nerve crush injury model. In vitro, the primary Schwann cells (SCs) and its cell line were cultured in normal medium containing NGF, their capable of swallowing or clearing degenerated myelin was evaluated through supplement of homogenized myelin fractions. Results: Administration of exogenous NGF could activate autophagy in dedifferentiated SCs, accelerate myelin debris clearance and phagocytosis, as well as promote axon and myelin regeneration at early stage of PNI. These NGF effects were effectively blocked by autophagy inhibitors. In addition, inhibition of the p75 kD neurotrophin receptor (p75NTR) signal or inactivation of the AMP-activated protein kinase (AMPK) also inhibited the NGF effect as well. Conclusions: NGF effect on promoting early nerve regeneration is closely associated with its accelerating autophagic clearance of myelin debris in SCs, which probably regulated by the p75NTR/AMPK/mTOR axis. Our studies thus provide strong support that NGF may serve as a powerful pharmacological therapy for peripheral nerve injuries.


Subject(s)
Autophagy/drug effects , Myelin Sheath/metabolism , Nerve Growth Factor/pharmacology , Nerve Regeneration/drug effects , Schwann Cells/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/pharmacology , Animals , Autophagy/physiology , Cell Line , Humans , Male , Mice , Mice, Knockout , Myelin Sheath/physiology , Nerve Growth Factor/administration & dosage , Nerve Growth Factor/metabolism , Nerve Regeneration/physiology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/pharmacology , Peripheral Nerve Injuries/metabolism , Phagocytosis/drug effects , Phagocytosis/physiology , Rats , Rats, Wistar , Receptors, Nerve Growth Factor/antagonists & inhibitors , Schwann Cells/physiology , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , TOR Serine-Threonine Kinases/metabolism
11.
Aging Dis ; 10(6): 1233-1245, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31788335

ABSTRACT

Autophagy is a lysosome-dependent cellular catabolic mechanism that mediates the turnover of dysfunctional organelles and aggregated proteins. It has a neuroprotective role on neurodegenerative diseases. Here, we hypothesized that autophagy may also have a neuroprotective role in diabetes-associated cognitive decline (DACD). In current study, we found that db/db mice display cognitive decline with inferior learning and memory function. The accumulation of ß-amyloid1-42 (Aß1-42), which is a characteristic of Alzheimer's disease (AD), was markedly higher in the prefrontal cortex (PFC), cornu ammon1 (CA1), and dentate gyrus (DG) areas of the hippocampus in db/db mice. Moreover, BDNF and microtubule associated protein 2 (MAP2) levels were lower in the hippocampus of db/db mice. However, there was no noticeable differences in the level of apoptosis in the hippocampus between control (CON) mice and db/db mice. Markers of autophagy in the hippocampus were elevated in db/db mice. The expression levels of ATG5, ATG7, and LC3B were higher, and the level of P62 was lower. An autophagy inhibitor, 3-MA, and ATG7 siRNA significantly reversed the activation of autophagy in vitro, which was accompanied with a higher level of apoptosis. Taken together, our current study suggests that diabetes is associated with cognitive decline, and activation of autophagy has a neuroprotective role in DACD.

12.
Am J Transl Res ; 9(2): 275-288, 2017.
Article in English | MEDLINE | ID: mdl-28337259

ABSTRACT

Brain injury secondary to birth asphyxia is the major cause of death and long-term disability in newborns. Intranasal drug administration enables agents to bypass the blood-brain barrier (BBB) and enter the brain directly. In this study, we determined whether intranasal basic fibroblast growth factor (bFGF) could exert neuroprotective effects in neonatal rats after hypoxic-ischaemic (HI) brain injury and assessed whether attenuation of endoplasmic reticulum (ER) stress was associated with these neuroprotective effects. Rats were subjected to HI brain injury via unilateral carotid artery ligation followed by 2.5 h of hypoxia and then treated with intranasal bFGF or vehicle immediately after HI injury. We found that the unfolded protein response (UPR) was strongly activated after HI injury and that bFGF significantly reduced the levels of the ER stress signalling proteins GRP78 and PDI. bFGF also decreased brain infarction volumes and conferred long-term neuroprotective effects against brain atrophy and neuron loss after HI brain injury. Taken together, our results suggest that intranasal bFGF provides neuroprotection function partly by inhibiting HI injury-induced ER stress. bFGF may have potential as a therapy for human neonates after birth asphyxia.

14.
Neurotherapeutics ; 13(4): 844-858, 2016 10.
Article in English | MEDLINE | ID: mdl-27170156

ABSTRACT

The blood-spinal cord barrier (BSCB) plays important roles in the recovery of spinal cord injury (SCI), and caveolin-1 is essential for the integrity and permeability of barriers. Basic fibroblast growth factor (bFGF) is an important neuroprotective protein and contributes to the survival of neuronal cells. This study was designed to investigate whether bFGF is beneficial for the maintenance of junction proteins and the integrity of the BSCB to identify the relations with caveolin-1 regulation. We examined the integrity of the BSCB with Evans blue dye and fluorescein isothiocyanate-dextran extravasation, measured the junction proteins and matrix metalloproteinases, and evaluated the locomotor function recovery. Our data indicated that bFGF treatment improved the recovery of BSCB and functional locomotion in contusive SCI model rats, reduced the expression and activation of matrix metalloproteinase-9, and increased the expressions of caveolin-1 and junction proteins, including occludin, claudin-5, p120-catenin, and ß-catenin. In the brain, in microvascular endothelial cells, bFGF treatment increased the levels of junction proteins, caveolin-1 small interfering RNA abolished the protective effect of bFGF under oxygen-glucose deprivation conditions, and the expression of fibroblast growth factor receptor 1 and co-localization with caveolin-1 decreased significantly, which could not be reversed by bFGF treatment. These findings provide a novel mechanism underlying the beneficial effects of bFGF on the BSCB and recovery of SCI, especially the regulation of caveolin-1.


Subject(s)
Blood-Brain Barrier/drug effects , Caveolin 1/metabolism , Fibroblast Growth Factor 2/therapeutic use , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Animals , Blood-Brain Barrier/physiology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Caveolin 1/genetics , Disease Models, Animal , Endothelial Cells/drug effects , Evans Blue/pharmacokinetics , Female , Gene Expression Regulation/drug effects , Hippocampus/cytology , Humans , Locomotion/drug effects , Microvessels/cytology , Neurons/drug effects , Neurons/physiology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley
15.
Am J Transl Res ; 8(4): 1864-75, 2016.
Article in English | MEDLINE | ID: mdl-27186310

ABSTRACT

This study aims to investigate the role of endocytoplasmic reticulum (ER) stress induced by spinal cord injury (SCI) in blood-spinal cord barrier (BSCB) disruption and the effect of phenylbutyrate (PBA) on BSCB disruption after SCI. After a moderate contusion injury at the T9 level of spinal cord with a vascular clip, PBA was immediately administered into injured rat via intraperitoneal injection (100 mg/kg) and then further treated once a day for 2 weeks for behavior test. Spinal cord was collected at 1 day post-injury for evaluation of the effects of ER stress and PBA on BSCB disruption after SCI. PBA significantly attenuated BSCB permeability and degradation of tight junction molecules such as P120, ß-catenin, Occludin and Claudin5 at 1 day after injury and improved functional recovery in the rat model of trauma. The BSCB protective effect of PBA is related to the inhibition of ER stress induced by SCI. In addition, PBA significantly inhibited the increase of ER stress markers and prevents loss of tight junction and adherens junction proteins in TG-treated human brain microvascular endothelial cells (HBMEC). Taken together, our data demonstrate that therapeutic strategies targeting ER stress may be suitable for the therapy of preserving BSCB integrity after SCI. PBA may be a new candidate as a therapeutic agent for protecting SCI by a compromised BSCB.

16.
Int Urol Nephrol ; 48(4): 547-52, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26897037

ABSTRACT

PURPOSE: Chronic kidney disease, characterized by gradual loss of renal function and irreversible progression, is becoming a major public health problem worldwide. Chronic kidney disease may lead to end-stage renal disease, as well as increase the morbidity and mortality associated with cardiovascular disease. METHODS: This review focuses on identifying risk factors indicating the need for intervention in early stages of chronic kidney disease, as well as determining factors that may improve patient prognosis. However, all the risk factors giving rise to the progression of chronic kidney disease have not yet been identified. RESULTS: Metabonomics is a new type of omics that reflects the real-time pathophysiology of disease and focuses on endogenous metabolites of low molecular weight. This new and powerful tool has recently been used to explore metabonomic biomarkers of chronic kidney disease, enabling early diagnosis and timely intervention and treatment to slow the progression of chronic kidney disease. CONCLUSIONS: This review summarizes recent findings on the identification, using a metabonomic approach, of biomarkers associated with risk factors for the development and progression of chronic kidney disease.


Subject(s)
Biomarkers/metabolism , Early Diagnosis , Metabolomics , Renal Insufficiency, Chronic/diagnosis , Disease Progression , Humans , Prognosis , Renal Insufficiency, Chronic/metabolism , Risk Factors
17.
Int J Biol Sci ; 12(1): 87-99, 2016.
Article in English | MEDLINE | ID: mdl-26722220

ABSTRACT

Spinal cord injury (SCI) induces the disruption of the blood-spinal cord barrier (BSCB) which leads to infiltration of blood cells, an inflammatory response, and neuronal cell death, resulting spinal cord secondary damage. Retinoic acid (RA) has a neuroprotective effect in both ischemic brain injury and SCI, however the relationship between BSCB disruption and RA in SCI is still unclear. In this study, we demonstrated that autophagy and ER stress are involved in the protective effect of RA on the BSCB. RA attenuated BSCB permeability and decreased the loss of tight junction (TJ) molecules such as P120, ß-catenin, Occludin and Claudin5 after injury in vivo as well as in Brain Microvascular Endothelial Cells (BMECs). Moreover, RA administration improved functional recovery in the rat model of SCI. RA inhibited the expression of CHOP and caspase-12 by induction of autophagic flux. However, RA had no significant effect on protein expression of GRP78 and PDI. Furthermore, combining RA with the autophagy inhibitor chloroquine (CQ) partially abolished its protective effect on the BSCB via exacerbated ER stress and subsequent loss of tight junctions. Taken together, the neuroprotective role of RA in recovery from SCI is related to prevention of of BSCB disruption via the activation of autophagic flux and the inhibition of ER stress-induced cell apoptosis. These findings lay the groundwork for future translational studies of RA for CNS diseases, especially those related to BSCB disruption.


Subject(s)
Apoptosis/drug effects , Blood-Brain Barrier/metabolism , Endoplasmic Reticulum Stress/physiology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Tretinoin/therapeutic use , Animals , Blood-Brain Barrier/drug effects , Cell Survival/drug effects , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Female , Humans , Rats , Rats, Sprague-Dawley
18.
J Cell Mol Med ; 20(6): 1062-75, 2016 06.
Article in English | MEDLINE | ID: mdl-26769343

ABSTRACT

After spinal cord injury (SCI), disruption of blood-spinal cord barrier (BSCB) elicits blood cell infiltration such as neutrophils and macrophages, contributing to permanent neurological disability. Previous studies show that epidermal growth factor (EGF) produces potent neuroprotective effects in SCI models. However, little is known that whether EGF contributes to the integrity of BSCB. The present study is performed to explore the mechanism of BSCB permeability changes which are induced by EGF treatment after SCI in rats. In this study, we demonstrate that EGF administration inhibits the disruption of BSCB permeability and improves the locomotor activity in SCI model rats. Inhibition of the PI3K/Akt pathways by a specific inhibitor, LY294002, suppresses EGF-induced Rac1 activation as well as tight junction (TJ) and adherens junction (AJ) expression. Furthermore, the protective effect of EGF on BSCB is related to the activation of Rac1 both in vivo and in vitro. Blockade of Rac1 activation with Rac1 siRNA downregulates EGF-induced TJ and AJ proteins expression in endothelial cells. Taken together, our results indicate that EGF treatment preserves BSCB integrity and improves functional recovery after SCI via PI3K-Akt-Rac1 signalling pathway.


Subject(s)
Epidermal Growth Factor/pharmacology , Epidermal Growth Factor/therapeutic use , Signal Transduction , Spinal Cord Injuries/blood , Spinal Cord Injuries/drug therapy , Spinal Cord/pathology , Adherens Junctions/drug effects , Adherens Junctions/metabolism , Animals , Chromones/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epidermal Growth Factor/administration & dosage , Female , Glucose/deficiency , Humans , Morpholines/pharmacology , Neuroprotective Agents/pharmacology , Oxygen , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proteolysis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Recovery of Function/drug effects , Signal Transduction/drug effects , Spinal Cord/drug effects , Tight Junctions/drug effects , Tight Junctions/metabolism , rac1 GTP-Binding Protein/metabolism
19.
Neurochem Res ; 41(4): 813-25, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26582233

ABSTRACT

Spinal cord injury (SCI) induces the disruption of the blood-spinal cord barrier (BSCB), which leads to infiltration of blood cells, inflammatory responses and neuronal cell death, with subsequent development of spinal cord secondary damage. Recent reports pointed to an important role of retinoic acid (RA), the active metabolite of the vitamin A, in the induction of the blood-brain barrier (BBB) during human and mouse development, however, it is unknown whether RA plays a role in maintaining BSCB integrity under the pathological conditions such as SCI. In this study, we investigated the BSCB protective role of RA both in vivo and in vitro and demonstrated that autophagy are involved in the BSCB protective effect of RA. Our data show that RA attenuated BSCB permeability and also attenuated the loss of tight junction molecules such as P120, ß-catenin, Occludin and Claudin5 after injury in vivo as well as in brain microvascular endothelial cells. In addition, RA administration improved functional recovery of the rat model of trauma. We also found that RA could significantly increase the expression of LC3-II and decrease the expression of p62 both in vivo and in vitro. Furthermore, combining RA with the autophagy inhibitor chloroquine (CQ) partially abolished its protective effect on the BSCB and exacerbated the loss of tight junctions. Together, our studies indicate that RA improved functional recovery in part by the prevention of BSCB disruption via the activation of autophagic flux after SCI.


Subject(s)
Autophagy , Spinal Cord Injuries/metabolism , Spinal Cord/blood supply , Tretinoin/pharmacology , Animals , Brain/blood supply , Catenins/metabolism , Cells, Cultured , Claudins/metabolism , Female , Humans , Microvessels/metabolism , Motor Activity/drug effects , Occludin/metabolism , Permeability , Rats, Sprague-Dawley , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , beta Catenin/metabolism , Delta Catenin
20.
Mol Neurobiol ; 53(10): 7298-7311, 2016 12.
Article in English | MEDLINE | ID: mdl-26687235

ABSTRACT

Many traumatic brain injury (TBI) survivors sustain neurological disability and cognitive impairments due to the lack of defined therapies to reduce TBI-induced blood-brain barrier (BBB) breakdown. Exogenous basic fibroblast growth factor (bFGF) has been shown to have neuroprotective function in brain injury. The present study therefore investigates the beneficial effects of bFGF on the BBB after TBI and the underlying mechanisms. In this study, we demonstrate that bFGF reduces neurofunctional deficits and preserves BBB integrity in a mouse model of TBI. bFGF suppresses RhoA and upregulates tight junction proteins, thereby mitigating BBB breakdown. In vitro, bFGF exerts a protective effect on BBB by upregulating tight junction proteins claudin-5, occludin, zonula occludens-1, p120-catenin, and ß-catenin under oxygen glucose deprivation/reoxygenation (OGD) in human brain microvascular endothelial cells (HBMECs). Both the in vivo and in vitro effects are related to the activation of the downstream signaling pathway, PI3K/Akt/Rac-1. Inhibition of the PI3K/Akt or Rac-1 by specific inhibitors LY294002 or si-Rac-1, respectively, partially reduces the protective effect of bFGF on BBB integrity. Overall, our results indicate that the protective role of bFGF on BBB involves the regulation of tight junction proteins and RhoA in the TBI model and OGD-induced HBMECs injury, and that activation of the PI3K/Akt /Rac-1 signaling pathway underlies these effects.


Subject(s)
Blood-Brain Barrier/pathology , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Fibroblast Growth Factor 2/pharmacology , Intercellular Junctions/metabolism , Membrane Proteins/metabolism , Signal Transduction/drug effects , Animals , Biomarkers/metabolism , Blood-Brain Barrier/injuries , Brain Injuries, Traumatic/pathology , Endothelial Cells/metabolism , Glucose/deficiency , Guanosine Triphosphate/metabolism , Humans , Intercellular Junctions/drug effects , Male , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxygen , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...