1.Protective effects of histone deacetylase 6 specific inhibitor tubastatin A on subarachnoid hemorrhage in rats and the underlying mechanisms.
Yuwei ZHU ; Haiping ZHENG ; Chunli CHEN
Journal of Central South University(Medical Sciences) 2023;48(2):172-181
		                        		
		                        			OBJECTIVES:
		                        			Subarachnoid hemorrhage (SAH) is a serious cerebrovascular disease. Early brain injury (EBI) and cerebral vasospasm are the main reasons for poor prognosis of SAH patients. The specific inhibitor of histone deacetylase 6 (HDAC6), tubastatin A (TubA), has been proved to have a definite neuroprotective effect on a variety of animal models of acute and chronic central nervous system diseases. However, the neuroprotective effect of TubA on SAH remains unclear. This study aims to investigate the expression and localization of HDAC6 in the early stage of SAH, and to evaluate the protective effects of TubA on EBI and cerebral vasospasm after SAH and the underlying mechanisms.
		                        		
		                        			METHODS:
		                        			Adult male SD rats were treated with modified internal carotid artery puncture to establish SAH model. In the first part of the experiment, rats were randomly divided into 6 groups: a sham group, a SAH-3 h group, a SAH-6 h group, a SAH-12 h group, a SAH-24 h group, and a SAH-48 h group. At 3, 6, 12, and 24 h after SAH modeling, the injured cerebral cortex of rats in each group was taken for Western blotting to detect the expression of HDAC6. In addition, the distribution of HDAC6 in the cerebral cortex of the injured side was measured by immunofluorescence double staining in SAH-24 h group rats. In the second part, rats were randomly divided into 4 groups: a sham group, a SAH group, a SAH+TubAL group (giving 25 mg/kg TubA), and a SAH+TubAH group (giving 40 mg/kg TubA). At 24 h after modeling, the injured cerebral cortex tissue was taken for Western blotting to detect the expression levels of HDAC6, endothelial nitric oxide synthase (eNOS), and inducible nitric oxide synthase (iNOS), terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) staining to detect apoptosis, and hematoxylin and eosin (HE) staining to detect the diameter of middle cerebral artery.
		                        		
		                        			RESULTS:
		                        			The protein expression of HDAC6 began to increase at 6 h after SAH (P<0.05), peaked at 24 h (P<0.001), and decreased at 48 h, but there was still a difference compared with the sham group (P<0.05). HDAC6 is mainly expressed in the cytoplasm of the neurons. Compared with the sham group, the neurological score was decreased significantly and brain water content was increased significantly in the SAH group (both P<0.01). Compared with the SAH group, the neurological score was increased significantly and brain water content was decreased significantly in the SAH+TubAH group (both P<0.05), while the improvement of the above indexes was not significant in the SAH+TubAL group (both P>0.05). Compared with the sham group, the expression of eNOS was significantly decreased (P<0.01) and the expressions of iNOS and HDAC6 were significantly increased (P<0.05 and P<0.01, respectively) in the SAH group. Compared with the SAH group, the expression of eNOS was significantly increased, and iNOS and HDAC6 were significantly decreased in the SAH+TubA group (all P<0.05). Compared with the SAH group, the number of TUNEL positive cells was significantly decreased and the diameter of middle cerebral artery was significantly increased in the SAH+TubA group (both P<0.05) .
		                        		
		                        			CONCLUSIONS
		                        			HDAC6 is mainly expressed in neurons and is up-regulated in the cerebral cortex at the early stage of SAH. TubA has protective effects on EBI and cerebral vasospasm in SAH rats by reducing brain edema and cell apoptosis in the early stage of SAH. In addition, its effect of reducing cerebral vasospasm may be related to regulating the expression of eNOS and iNOS.
		                        		
		                        		
		                        		
		                        			Rats
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Rats, Sprague-Dawley
		                        			;
		                        		
		                        			Subarachnoid Hemorrhage/drug therapy*
		                        			;
		                        		
		                        			Vasospasm, Intracranial/metabolism*
		                        			;
		                        		
		                        			Histone Deacetylase Inhibitors/therapeutic use*
		                        			;
		                        		
		                        			Neuroprotective Agents/therapeutic use*
		                        			;
		                        		
		                        			Histone Deacetylase 6/pharmacology*
		                        			;
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			Brain Injuries/drug therapy*
		                        			
		                        		
		                        	
2.Effects of cysteinyl leukotrienes receptor antagonists on chronic brain injury after global cerebral ischemia/reperfusion.
Hao WANG ; Honggang GUO ; Qi LOU ; Qiaojuan SHI
Journal of Zhejiang University. Medical sciences 2018;47(1):19-26
		                        		
		                        			OBJECTIVE:
		                        			: To investigate the effects of cysteinyl leukotrienes receptor (CysLTR) antagonists on global cerebral ischemia/reperfusion (CI/R) injury in gerbils, and to explore its mechanism.
		                        		
		                        			METHODS:
		                        			: Totally 40 gerbils weighting 45-65 g were randomized into sham, saline, Pranlukast and HAMI 3379 groups with 10 animals in each. The CI/R model was established in gerbils by bilateral common carotid occlusion for 10 min followed by reperfusion. After ischemia, the CysLTR antagonists Pranlukast (0.1 mg/kg) and HAMI 3379 (0.1 mg/kg) were injected intraperitoneally for 5 consecutive days in the last two groups,while the former two groups were injected with saline only (10 mL/kg). After 24 h or 14 d reperfusion, neurological deficit score was evaluated and the behavioral dysfunction was assessed, respectively. And 14 d after reperfusion, the neuron morphology of cerebral cortex was observed in brain sections stained with Cresyl violet. In addition, the Iba-1 (microgila) and GFAP (astrocyte) positive cells in cerebral cortex were observed by using immunohistochemitry method.
		                        		
		                        			RESULTS:
		                        			: CI/R models were successfully established in 21 out of 30 gerbils with 7 in saline group, 6 in Pranlukast group, and 8 in HAMI 3379 group. Compared with saline group, Pranlukast and HAMI 3379 significantly attenuated neurological deficits, improved the behavioral function 24 h after reperfusion(all <0.01); Pranlukast and HAMI 3379 also significantly improved the behavioral function 14 days after reperfusion(<0.05 or <0.01). Compared with saline group, the neurological symptom scores in Pranlukast and HAMI 3379 groups presented a trend of amelioration 14 d after reperfusion, but it was not significant(>0.05). In addition, Pranlukast and HAMI 3379 also inhibited the neuron loss and injury, suppressed microgila and astrocyte activation 14 d after reperfusion(all <0.01).
		                        		
		                        			CONCLUSIONS
		                        			: CysLTR antagonists Pranlukast and HAMI 3379 have long-term neuroprotective effect on chronic brain injury induced by global cerebral ischemia/reperfusion in gerbils.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Behavior, Animal
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Brain Injury, Chronic
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			Brain Ischemia
		                        			;
		                        		
		                        			Gerbillinae
		                        			;
		                        		
		                        			Leukotriene Antagonists
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Neuroprotective Agents
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Random Allocation
		                        			;
		                        		
		                        			Receptors, Leukotriene
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Reperfusion Injury
		                        			;
		                        		
		                        			drug therapy
		                        			
		                        		
		                        	
3.Neuroprotective effect of rapamycin against Parkinson's disease in mice.
Feng ZHU ; Miao FAN ; Ziwei XU ; Yiting CAI ; Yizhen CHEN ; Shuang YU ; Linghui ZENG
Journal of Zhejiang University. Medical sciences 2018;47(5):465-472
		                        		
		                        			OBJECTIVE:
		                        			To investigate the effect of rapamycin on Parkinson's disease (PD) and its underlying mechanism in mice.
		                        		
		                        			METHODS:
		                        			Sixty SPF adult male C57BL/6 mice were randomly divided into control group, model group and treatment group. 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine(MPTP) was used to induce Parkinson's disease in model group and treatment group. All mice were trained to cross the runway and were subjected to computer-assisted CatWalk. The numbers of tyrosine hydroxylase positive (TH) neurons in the substantia nigra (SN) were assessed by unbiased stereology using the optical fractionator method; protein expression was detected by Western blot analysis; and glutathione peroxidase (GSH-Px), malondialdehyde (MDA) and superoxide dismutase (SOD) were detected by spectrophotometry.
		                        		
		                        			RESULTS:
		                        			In the model group, a decrease in stride rate and an increase in variation of stance and swing were noted by CatWalk system (<0.05 or <0.01); the numbers of TH neurons decreased (<0.01); expression of p-Akt, p-S6K, p-S6 and p-ULK increased (all <0.01); LC3-Ⅱ/Ⅰ ratio decreased (<0.01); MDA level was elevated while the levels of SOD and GSH-PX were reduced (all <0.01). Compared with the model group, after treated with rapamycin, the abnormal behavior including the stride length, variation of stance and swing and step patterns induced by MPTP were all improved (<0.05 or <0.01); the numbers of TH neurons increased (<0.05); the expression of p-Akt, p-S6K, p-S6 and p-ULK was suppressed (all <0.01); the LC3-Ⅱ/Ⅰ ratio was upregulated (<0.05); MDA level decreased while the levels of GSH-Px and SOD increased (all <0.01).
		                        		
		                        			CONCLUSIONS
		                        			Rapamycin inhibits the activation of mTOR pathway, which contributes to protect against the loss of dopaminergic neurons and provide behavioral improvements in mice with Parkinson's disease. These results are partially related to the ability of rapamycin in inducing autophagy and reducing oxidative stress.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Behavior, Animal
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Disease Models, Animal
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Mice, Inbred C57BL
		                        			;
		                        		
		                        			Neuroprotective Agents
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Oxidative Stress
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Parkinson Disease
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			Random Allocation
		                        			;
		                        		
		                        			Sirolimus
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Substantia Nigra
		                        			;
		                        		
		                        			drug effects
		                        			
		                        		
		                        	
4.Fluoxetine is Neuroprotective in Early Brain Injury via its Anti-inflammatory and Anti-apoptotic Effects in a Rat Experimental Subarachnoid Hemorrhage Model.
Hui-Min HU ; Bin LI ; Xiao-Dong WANG ; Yun-Shan GUO ; Hua HUI ; Hai-Ping ZHANG ; Biao WANG ; Da-Geng HUANG ; Ding-Jun HAO
Neuroscience Bulletin 2018;34(6):951-962
		                        		
		                        			
		                        			Fluoxetine, an anti-depressant drug, has recently been shown to provide neuroprotection in central nervous system injury, but its roles in subarachnoid hemorrhage (SAH) remain unclear. In this study, we aimed to evaluate whether fluoxetine attenuates early brain injury (EBI) after SAH. We demonstrated that intraperitoneal injection of fluoxetine (10 mg/kg per day) significantly attenuated brain edema and blood-brain barrier (BBB) disruption, microglial activation, and neuronal apoptosis in EBI after experimental SAH, as evidenced by the reduction of brain water content and Evans blue dye extravasation, prevention of disruption of the tight junction proteins zonula occludens-1, claudin-5, and occludin, a decrease of cells staining positive for Iba-1, ED-1, and TUNEL and a decline in IL-1β, IL-6, TNF-α, MDA, 3-nitrotyrosine, and 8-OHDG levels. Moreover, fluoxetine significantly improved the neurological deficits of EBI and long-term sensorimotor behavioral deficits following SAH in a rat model. These results indicated that fluoxetine has a neuroprotective effect after experimental SAH.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Blood-Brain Barrier
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Brain Edema
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			etiology
		                        			;
		                        		
		                        			Cytokines
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Disease Models, Animal
		                        			;
		                        		
		                        			Fluoxetine
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			In Situ Nick-End Labeling
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Neuroprotective Agents
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Pain Measurement
		                        			;
		                        		
		                        			Psychomotor Performance
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			RNA, Messenger
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Rats
		                        			;
		                        		
		                        			Rats, Sprague-Dawley
		                        			;
		                        		
		                        			Subarachnoid Hemorrhage
		                        			;
		                        		
		                        			complications
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Time Factors
		                        			;
		                        		
		                        			Vasospasm, Intracranial
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			etiology
		                        			
		                        		
		                        	
5.Angelica tenuissima Nakai Ameliorates Cognitive Impairment and Promotes Neurogenesis in Mouse Model of Alzheimer's Disease.
Minji CHOI ; Younghyurk LEE ; Seung-Hun CHO
Chinese journal of integrative medicine 2018;24(5):378-384
OBJECTIVETo research Angelica tenuissima Nakai (ATN) for use in novel Alzheimer's disease (AD) therapeutics.
METHODSThe effect of a 30% ethanol extract of ATN (KH032) on AD-like cognitive impairment and neuropathological and neuroinflammatory changes induced by bilateral intracerebroventricular injections of β-amyloid (Aβ) peptide (Aβ) was investigated. Male C57Bl/6 mice were randomly divided into 4 groups, 10 in each group. KH032-treated groups were administrated with a low or high dose of KH032 (50 and 200 mg/kg, respectively), intragastrically for 16 days; distilled water was applied in the sham and negative groups. Open fifield test, Y maze and Morris water maze test were used for behavior test and cognitive ability. In addition, the neuroprotective effects of KH032 in Aβ-infused mice on the histopathological markers [neuronspecific nuclear protein (NeuN), Aβ] of neurodegeneration were examined. The levels of glial fibrillary acidic protein (GFAP), NeuN, phosphorylation extracellular signal-regulated kinase (ERK)/ERK, brain-derived neurotrophic factor (BDNF), phosphorylation cAMP response element-binding (CREB)/CREB protein expression were measured by Western blot.
RESULTSKH032 treatment ameliorated cognitive impairments, reduced the overexpression of Aβ, and inhibited neuronal loss and neuroinflammatory response in the Aβ-infused mice. Moreover, KH032 treatment enhanced BDNF expression levels in the hippocampus. Finally, KH032 treatment increased phosphorylation of ERK1/2 and CREB, vital for ERK-CREB signaling.
CONCLUSIONSKH032 attenuated cognitive defificits in the Aβ-infused mice by increasing BDNF expression and ERK1/2 and CREB phosphorylation and inhibiting neuronal loss and neuroinflflammatory response, suggesting that KH032 has therapeutic potential in neurodegenerative disorders such as AD.
Alzheimer Disease ; drug therapy ; pathology ; physiopathology ; Amyloid beta-Peptides ; Angelica ; chemistry ; Animals ; Brain ; pathology ; Brain-Derived Neurotrophic Factor ; metabolism ; Cognitive Dysfunction ; complications ; drug therapy ; physiopathology ; Cyclic AMP Response Element-Binding Protein ; metabolism ; Disease Models, Animal ; Male ; Maze Learning ; drug effects ; Memory, Short-Term ; drug effects ; Mice, Inbred C57BL ; Neurogenesis ; drug effects ; Neuroglia ; drug effects ; metabolism ; pathology ; Neurons ; drug effects ; metabolism ; pathology ; Neuroprotective Agents ; pharmacology ; therapeutic use ; Phosphorylation ; drug effects ; Phytotherapy ; Plant Extracts ; pharmacology ; therapeutic use ; Plaque, Amyloid ; drug therapy ; pathology ; physiopathology ; Signal Transduction ; drug effects
6.Neuroprotective effect of peptides extracted from walnut (Juglans Sigilata Dode) proteins on Aβ25-35-induced memory impairment in mice.
Juan ZOU ; Pei-shan CAI ; Chao-mei XIONG ; Jin-lan RUAN
Journal of Huazhong University of Science and Technology (Medical Sciences) 2016;36(1):21-30
		                        		
		                        			
		                        			Alzheimer's disease (AD) is one of the major neurodegenerative disorders of the elderly, which is characterized by the accumulation and deposition of amyloid-beta (Aβ) peptide in human brains. Oxidative stress and neuroinflammation induced by Aβ in brain are increasingly considered to be responsible for the pathogenesis of AD. The present study aimed to determine the protective effects of walnut peptides against the neurotoxicity induced by Aβ25-35 in vivo. Briefly, the AD model was induced by injecting Aβ25-35 into bilateral hippocampi of mice. The animals were treated with distilled water or walnut peptides (200, 400 and 800 mg/kg, p.o.) for five consecutive weeks. Spatial learning and memory abilities of mice were investigated by Morris water maze test and step-down avoidance test. To further explore the underlying mechanisms of the neuroprotectivity of walnut peptides, the activities of superoxide dismutase (SOD), glutathione (GSH), acetylcholine esterase (AChE), and the content of malondialdehyde (MDA) as well as the level of nitric oxide (NO) in the hippocampus of mice were measured by spectrophotometric method. In addition, the levels of 8-hydroxy-2'-deoxyguanosine (8-OHdG), tumor necrosis factor-α (TNF-α), interleukin 1β (IL-1β) and IL-6 in the samples were determined using ELISA. The hippocampal expressions of inducible nitric oxide synthase (iNOS) and nuclear factor κB (NF-κB) were evaluated by Western blot analysis. The results showed that walnut peptides supplementation effectively ameliorated the cognitive deficits and memory impairment of mice. Meanwhile, our study also revealed effective restoration of levels of antioxidant enzymes as well as inflammatory mediators with supplementation of walnut peptides (400 or 800 mg/kg). All the above findings suggested that walnut peptides may have a protective effect on AD by reducing inflammatory responses and modulating antioxidant system.
		                        		
		                        		
		                        		
		                        			Acetylcholinesterase
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Alzheimer Disease
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			etiology
		                        			;
		                        		
		                        			Amyloid beta-Peptides
		                        			;
		                        		
		                        			toxicity
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Glutathione
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Hippocampus
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Interleukins
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Juglans
		                        			;
		                        		
		                        			chemistry
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Malondialdehyde
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Maze Learning
		                        			;
		                        		
		                        			Memory Disorders
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			etiology
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			NF-kappa B
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Neuroprotective Agents
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Nitric Oxide
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Peptide Fragments
		                        			;
		                        		
		                        			toxicity
		                        			;
		                        		
		                        			Peptides
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Plant Extracts
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Superoxide Dismutase
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Tumor Necrosis Factor-alpha
		                        			;
		                        		
		                        			metabolism
		                        			
		                        		
		                        	
7.Adenosine Al Receptor Mediated Neuroprotection of Shenmai Injection on Rat Cerebral Ischemia/Reperfusion Injury: an Experimental Study.
Hua-rong LU ; Sheng-wen SONG ; Kun-yuan HAN ; Hai-peng LIU ; Shuang-dong CHEN ; Jun-lu WANG ; Qin-xue DAI
Chinese Journal of Integrated Traditional and Western Medicine 2015;35(9):1109-1112
OBJECTIVETo observe whether adenosine Al receptor (Al R) mediated neuroprotection of Shenmai Injection (SI) on rat cerebral ischemia/reperfusion (I/R) injury.
METHODSThe focal cerebral I/R model was established by middle cerebral artery occlusion (MCAO). Totally 60 successfully modeled rats was divided into 5 groups according to randomized block principle, i.e., the model group, the SI group, the SI + AlR antagonist (1,3-dipropyl-8-cyclopentylxanthine, DPCPX) group, the AlR antagonist control group, and the dimethyl sulfoxide (DMSO) control group, 12 in each group. Besides, a sham-operation group was set up (n =12). SI at 15 mL/kg was peritoneally injected to mice in the SI group immediately after cerebral I/R. Equal volume of normal saline was injected to mice in the model group and the sham-operation group. DPCPX at 1 mg/mL was peritoneally injected to mice in the Al R antagonist control group 30 min before peritoneal injecting SI. DPCPX at 1 mg/kg and DMSO at 1 mL/kg were peritoneally injected to mice in the AlR antagonist control group and the DMSO control group 30 min immediately before cerebral I/R. Rats' neurobehavioral scores were assessed after 24 h reperfusion. The volume of cerebral infarction and Bcl-2 protein expression of cerebral infarction penumbra were also detected. Results Compared with the sham-operation group, neurobehavioral scores, the volume of cerebral infarction, and Bcl-2 protein expression increased (all P <0. 05). Compared with the model group, neurobehavioral scores and the volume of cerebral infarction obviously decreased, but Bcl-2 protein expression increased in the SI group (all P <0. 05). Compared with the SI group, neurobehavioral scores increased, the volume of cerebral infarction was obviously enlarged, and Bcl-2 protein expression was obviously reduced in the A1R antagonist control group (all P <0. 05).
CONCLUSIONSSI's neurobehavioral scores could be partially reversed in the Al R antagonist control group, the volume of cerebral infarction and Bcl-2 protein expression improved. AlR might possibly meditate neuroprotection of SI on MACO mire
Adenosine ; Animals ; Brain Ischemia ; drug therapy ; Drug Combinations ; Drugs, Chinese Herbal ; pharmacology ; therapeutic use ; Infarction, Middle Cerebral Artery ; Mice ; Neuroprotection ; physiology ; Neuroprotective Agents ; pharmacology ; therapeutic use ; Rats ; Rats, Sprague-Dawley ; Receptor, Adenosine A1 ; metabolism ; Reperfusion Injury ; drug therapy ; Xanthines
8.Neuroprotective Effect of Compound Anisodine in a Mouse Model with Chronic Ocular Hypertension.
Wen-Dong LIU ; Lan-Lan CHEN ; Ce-Ying SHEN ; Li-Bin JIANG
Chinese Medical Journal 2015;128(19):2652-2657
BACKGROUNDCompound anisodine (CA) is a compound preparation made from hydrobromide anisodine and procaine hydrochloride. The former is an M-choline receptor blocker with the function of regulating the vegetative nervous system, improving microcirculation, and so on. The latter is an antioxidant with the activities of neuroprotection. This study aimed to investigate the potential neuroprotection of CA, which affects the degeneration of the retinal ganglion cells (RGCs) in an animal model with chronic ocular hypertension.
METHODSFemale C57BL/6J mice (n = 24) were divided randomly into four groups: normal control group without any treatment (Group A, n = 6); CA control group with feeding the CA solution (Group B, n = 6); microbeads (MBs) control group with injecting MB into the anterior chamber (Group C, n = 6); CA study group with MB injection and with feeding the CA solution (Group D, n = 6). Intraocular pressure (IOP) was measured every 3 days after MB injection. At the 21st day, neurons were retrograde-labeled by Fluoro-Gold (FG). Animals were sacrificed on the 27th day. Retinal flat mounts were stained immunohistologically by α2-III-tubulin. FG-retrograde-labeled RGCs, α2-III-tubulin-positive RGCs, and α2-III-tubulin-positive nerve fibers were quantified.
RESULTSMice of Groups C and D expressed the incidence of consistent IOP elevation, which is above the IOP level of Group A with the normal one. There is no significant difference in IOP between Groups A and B (P > 0.05). On the 27th day, there were distinct loss in stained RGCs and nerve fibers from Groups C and D compared with Group A (allP < 0.001). The quantity was significantly higher in Group D as compared to Group C (allP < 0.001) but lower than Group A (allP > 0.001). There was no significant difference in the quantity of RGCs and nerve fibers between Groups A and B (allP > 0.05).
CONCLUSIONSThese findings suggest that CA plays an importantly neuroprotective role on RGCs in a mouse model with chronic ocular hypertension.
Animals ; Cell Survival ; drug effects ; Female ; Immunohistochemistry ; Intraocular Pressure ; drug effects ; Mice ; Mice, Inbred C57BL ; Neuroprotective Agents ; pharmacology ; therapeutic use ; Ocular Hypertension ; drug therapy ; Random Allocation ; Retinal Ganglion Cells ; drug effects ; Scopolamine Derivatives ; pharmacology ; therapeutic use
9.Effect of glycyrrhizin on traumatic brain injury in rats and its mechanism.
Xiang-Jin GU ; Jin XU ; Ban-You MA ; Gong CHEN ; Pei-Yuan GU ; Dong WEI ; Wei-Xing HU
Chinese Journal of Traumatology 2014;17(1):1-7
OBJECTIVETo investigate the neuroprotective effects of glycyrrhizin (Gly) as well as its effect on expression of high-mobility group box 1 (HMGB1) in rats after traumatic brain injury (TBI).
METHODSMale Sprague-Dawley rats were randomly divided into three groups: sham group, TBI group, and TBI+Gly group (n=36 per group). Rat TBI model was made by using the modified Feeney's method. In TBI+Gly group, Gly was administered intravenously at a dosage of 10 mg/kg 30 min after TBI. At 24 h after TBI, motor function and brain water content were evaluated. Meanwhile, HMGB1/HMGB1 receptors including toll-like receptor 4 (TLR4) and receptor for advanced glycation end products (RAGE)/nuclear factor-κB(NF-κB) signaling pathway and inflammatory cytokines in the injured brain tissues were detected using quantitative real-time polymerase chain reaction, western blot, electrophoretic mobility shift assay and enzyme-linked immunosorbent assay. Furthermore, HMGB1, RAGE and TLR4 immunohistochemistry and apoptosis were analyzed.
RESULTSBeam walking performance impairment and brain edema were significantly reduced in TBI+Gly group compared with TBI group; meanwhile, the over-expressions of HMGB1/HMGB1 receptors (TLR4 and RAGE)/NF-κB DNA-binding activity and inflammatory cytokines were inhibited. The percentages of HMGB1, RAGE and TLR4-positive cells and apoptotic cells were respectively 58.37% ± 5.06%, 54.15% ± 4.65%, 65.50% ± 4.83%, 52.02% ± 4.63% in TBI group and 39.99% ± 4.99%, 34.87% ± 5.02%, 43.33% ± 4.54%, 37.84% ± 5.16% in TBI+Gly group (all P<0.01 compared with TBI group).
CONCLUSIONGly can reduce secondary brain injury and improve outcomes in rat following TBI by down-regulation of HMGB1/HMGB1 receptors (TLR4 and RAGE)/NF-κB-mediated inflammatory responses in the injured rat brain.
Animals ; Brain Injuries ; drug therapy ; Glycyrrhizic Acid ; pharmacology ; therapeutic use ; HMGB1 Protein ; metabolism ; Male ; Neuroprotective Agents ; therapeutic use ; Rats ; Rats, Sprague-Dawley
10.Progress on the pharmacological research of puerarin: a review.
Shu-Yong WEI ; Yi CHEN ; Xiao-Yu XU
Chinese Journal of Natural Medicines (English Ed.) 2014;12(6):407-414
		                        		
		                        			
		                        			Contemporary pharmacological research has demonstrated that puerarin, the most important phytoestrogen extracted from Pueraria lobata(Willd.) Ohwi, has protecting functions on the cardiovascular system, nervous system, osteoporosis, liver injury, and inflammation in vivo and in vitro. Most of these research studies focused on inhibiting oxidative stress and apoptosis through regulating various bioactivators and signal pathways. Among these, superoxide dismutase (SOD), endothelial nitric oxide synthase (eNOS) and malondialdehyde (MDA), and PI3K/Akt, MAPK, and NF-κB are of great importance. The data cited in this review were mainly obtained from articles listed in PubMed and Elsevier SDOL published from 1959 to 2013, and the search term used was "puerarin".
		                        		
		                        		
		                        		
		                        			Anti-Inflammatory Agents
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Antioxidants
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cardiovascular Diseases
		                        			;
		                        		
		                        			prevention & control
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Isoflavones
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Liver Diseases
		                        			;
		                        		
		                        			prevention & control
		                        			;
		                        		
		                        			Neuroprotective Agents
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Oxidative Stress
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Phytoestrogens
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Phytotherapy
		                        			;
		                        		
		                        			Plant Extracts
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Pueraria
		                        			;
		                        		
		                        			chemistry
		                        			
		                        		
		                        	
            
Result Analysis
Print
Save
E-mail