1.Resveratrol improves cognitive function in severely burned rats by inhibiting hippocampal NF-κB/JNK pathway.
Liang XING ; Wei WEI ; Cuina ZHANG ; Bingquan GAO ; Jianke FENG ; Leilei MA
Chinese Journal of Cellular and Molecular Immunology 2023;39(7):604-609
		                        		
		                        			
		                        			Objective To investigate the protective effect of resveratrol (RSV) on improving cognitive function in severely burned rats and its possible mechanism. Methods 18 male SD rats aged 18-20 months were randomly divided into 3 groups: control group, model group and RSV group, with 6 rats in each group. After successful modeling, the rats in RSV group were gavaged once daily with RSV (20 mg/kg). Meanwhile, the rats in control group and model group were gavaged once daily with an equal volume of sodium chloride solution. After 4 weeks, the cognitive function of all rats was estimated by Step-down Test. The concentration of tumor necrosis factor α (TNF-α) and interleukin 6 (IL-6) protein in serum of rats were detected by ELISA. The expression of IL-6, TNF-α mRNA and protein were estimated by real-time PCR and Western blotting. The apoptosis of hippocampal neurons was tested by terminal deoxynuclectidyl transferase-mediated dUTP-biotin nick end labeling assay (TUNEL). The expression of nuclear transcription factor-κB (NF-κB)/c-Jun N-terminal kinase (JNK) pathway-related proteins in hippocampus were assessed by Western blotting. Results Compared with the rats in model group, rats in RSV group exhibited improved cognitive function. Consistently, the rats in RSV group had a reduced concentration of TNF-α and IL-6 in serum, decreased mRNA and protein expressions of TNF-α and IL-6 in hippocampus, and decreased apoptosis rate and relative expression of p-NF-κB p65/NF-κB p65 and p-JNK/JNK in hippocampal neurons. Conclusion RSV alleviates inflammatory response and hippocampal neuronal apoptosis by inhibiting NF-κB/JNK pathway, thereby improving cognitive function in severely burned rats.
		                        		
		                        		
		                        		
		                        			Resveratrol/pharmacology*
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Rats
		                        			;
		                        		
		                        			Rats, Sprague-Dawley
		                        			;
		                        		
		                        			Burns/drug therapy*
		                        			;
		                        		
		                        			Cognition/drug effects*
		                        			;
		                        		
		                        			Hippocampus/metabolism*
		                        			;
		                        		
		                        			MAP Kinase Signaling System
		                        			;
		                        		
		                        			NF-kappa B/metabolism*
		                        			;
		                        		
		                        			Tumor Necrosis Factor-alpha/blood*
		                        			;
		                        		
		                        			Interleukin-6/blood*
		                        			;
		                        		
		                        			Neurons/drug effects*
		                        			;
		                        		
		                        			Apoptosis
		                        			
		                        		
		                        	
3.Improved effects of saponins from Panax japonicus on decline of cognitive function in natural aging rats via NLRP3 inflammasome pathway.
Bo RUAN ; Rui WANG ; Yuan-Jian YANG ; Dong-Fan WANG ; Jia-Wen WANG ; Chang-Cheng ZHANG ; Ding YUAN ; Zhi-Yong ZHOU ; Ting WANG
China Journal of Chinese Materia Medica 2019;44(2):344-349
		                        		
		                        			
		                        			The aim of this paper was to investigate the effect of total saponins from Panax japonicus( SPJ) on cognitive decline of natural aging rats and its mechanism. Thirty male SD rats of eighteen month old were randomly divided into three groups: aged group,10 mg·kg~(-1) SPJ-treated group and 30 mg·kg~(-1) SPJ-treated group. The SPJ-treated groups were given SPJ at the dosages of 10 mg·kg~(-1) and 30 mg·kg~(-1),respectively,from the age of 18 to 24 months. Aged group were lavaged the same amount of saline,10 six-month-old rats were used as control group,with 10 rats in each group. The open field test,novel object recognition and Morris water maze were performed to detect the changes of cognitive function in each group. The changes of synaptic transmission of long-term potentiation( LTP) in hippocampal CA1 region were detected by field potential recording. Western blot was used to detect the protein levels of NLRP3,ASC,caspase-1 and the changes of Glu A1,Glu A2,CAMKⅡ,CREB and phosphorylation of CAMKⅡ,CREB in each group.The results showed that SPJ could improve the decline of cognitive function in aging rats,reduce the damage of LTP in the hippocampal CA1 region of aged rats,and decrease the expression of NLRP3,ASC,caspase-1 in aging rats. At the same time,SPJ could enhance the membrane expression of AMPA receptor( Glu A1 and Glu A2),and increase the expression of p-CAMKⅡand p-CREB in aging rats.SPJ could improve cognitive decline of natural aging rats,and its mechanism may be related to regulating NLRP3 inflammasome,thus regulating the membrane expression of AMPA receptor,and enhancing the expression phosphorylation of CAMKⅡ and CREB.
		                        		
		                        		
		                        		
		                        			Aging
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			CA1 Region, Hippocampal
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Cognition
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Inflammasomes
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Long-Term Potentiation
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			NLR Family, Pyrin Domain-Containing 3 Protein
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Panax
		                        			;
		                        		
		                        			chemistry
		                        			;
		                        		
		                        			Random Allocation
		                        			;
		                        		
		                        			Rats
		                        			;
		                        		
		                        			Rats, Sprague-Dawley
		                        			;
		                        		
		                        			Saponins
		                        			;
		                        		
		                        			pharmacology
		                        			
		                        		
		                        	
4.Effect of N-acetylcysteine on Cognitive Function and Nuclear Factor Erythroid 2 Related Factor 2/Heme Oxygenase-1 Pathway in Mouse Models of Postoperative Cognitive Dysfunction.
Acta Academiae Medicinae Sinicae 2019;41(4):529-535
		                        		
		                        			
		                        			To investigate the effect of N-acetylcysteine(NAC)on cognitive function and nuclear factor erythroid 2 related factor 2/ heme oxygenase-1(Nrf2/HO-1)pathway in mouse models of postoperative cognitive dysfunction. Methods Fifty-four male C57BL/6J mice(3-4 months old)were randomly divided into control group,surgery group,and surgery+NAC group by block randomization.The intramedullary fixation for left tibial fracture surgery was performed to establish postoperative cognitive dysfunction models.NAC(150 mg/kg)was administered intraperitoneally in group surgery+NAC 30 minutes before and 3 hours,6 hours after surgery,while saline was given in control group and surgery group.Six mice in each group were selected randomly underwent Morris water maze test on the third day after surgery.Animals were sacrificed at the first and third postoperative days,and the hippocampus was harvested.Enzyme-linked immunosorbent assay was used to quantify the levels of interleukin-6(IL-6)and malondialdehyde(MDA)in hippocampus.Western blot and real-time polymerase chain reaction were used to measure the expressions of Nrf2 and HO-1 in hippocampus. Results There was no significant difference in swimming speed among three groups(=2.135,=0.114).Compared with control group and surgery+NAC group,the surgery group had prolonged escape latency(<0.01),reduced platform crossing times(<0.01),and shortened time spent in the target quadrant(<0.01).Compared with the control group,the surgery group and the surgery+NAC group had significantly increased levels of IL-6 and MDA in hippocampus at the first postoperative day(all =0.000).On the third postoperative day,there was no significant difference in the levels of IL-6(=0.251)and MDA(=0.103)between control group and surgery+NAC group.The protein expressions of Nrf2 and HO-1 in hippocampus were significantly higher in surgery group and surgery+NAC group than in control group and significantly higher in surgery+NAC group than in surgery group(all =0.000).The mRNA expressions of Nrf2 and HO-1 in hippocampus were significantly higher in surgery group and surgery+NAC group than in control group and significantly higher in surgery+NAC group than in surgery group (all =0.000). Conclusions NAC pretreatment may reduce oxidative stress and inflammatory response in hippocampus and improve cognitive function.Such effect may be relate to the activation of Nrf2/HO-1 pathway.
		                        		
		                        		
		                        		
		                        			Acetylcysteine
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Cognition
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cognitive Dysfunction
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			etiology
		                        			;
		                        		
		                        			Heme Oxygenase-1
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Membrane Proteins
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Mice, Inbred C57BL
		                        			;
		                        		
		                        			NF-E2-Related Factor 2
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Postoperative Complications
		                        			;
		                        		
		                        			Random Allocation
		                        			
		                        		
		                        	
5.Influence of general anesthetic exposure in developing brain on cognition and the underlying mechanisms.
Xin ZHAO ; Li-Jun HAO ; Yu-Tong ZHANG ; Yu ZHANG ; Ce ZHANG
Acta Physiologica Sinica 2019;71(5):749-759
		                        		
		                        			
		                        			With the evolution of medical techniques and technology, an increasing number of infants, neonates, and fetuses are exposed to general anesthesia for clinical diagnostic and therapeutic process. The neurotoxic effects of general anesthetics on developing brain have been a subject of concern and considerable research interest. Population-based study confirmed that single short-term general anesthetic exposure does not affect nervous system function, but multiple exposures to general anesthesia could damage cognitive function. Animal studies further discovered the underlying mechanisms. Nervous system is most susceptible to general anesthetics during the brain growth spurt. The time-point is more critical than the duration of exposure to general anesthetics. General anesthetics can induce intracellular calcium overload, disturb energy metabolism, promote cell apoptosis and lead to cell loss. General anesthetics can damage synaptic structure, transmission and plasticity, and impair brain function. High throughput omics technologies have been used to screen the differentially expressed genes induced by general anesthetics, which provide further understanding of the mechanism of general anesthetics affecting cognitive function. This review provides an update on the pathophysiologic mechanisms underlying the anesthesia-neurotoxicity, which will be helpful to provide instructions for the clinical use of general anesthesia in children.
		                        		
		                        		
		                        		
		                        			Anesthesia, General
		                        			;
		                        		
		                        			adverse effects
		                        			;
		                        		
		                        			Anesthetics, General
		                        			;
		                        		
		                        			adverse effects
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Brain
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			growth & development
		                        			;
		                        		
		                        			Cognition
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Infant
		                        			;
		                        		
		                        			Infant, Newborn
		                        			;
		                        		
		                        			Pregnancy
		                        			;
		                        		
		                        			Prenatal Exposure Delayed Effects
		                        			;
		                        		
		                        			physiopathology
		                        			
		                        		
		                        	
6.The effects of anabolic-androgenic steroids on behavioral, cognitive functions and nervous systems of adolescents.
Jia-Min WU ; Ying-Yi DENG ; Chu-Qian WEI ; Jin-Hong YAN
Acta Physiologica Sinica 2019;71(3):463-470
		                        		
		                        			
		                        			Anabolic-androgenic steroid (AAS) is responsible for muscle building and masculinizing. Using AAS can enhance muscle development and strength, and improve athletic performance. AAS abuse is not only seen in sport. Research has shown that there is an increasing number of adolescent AAS abusers. Adolescents are at a critical period of physical and mental development. Sex hormones are one of the important physiological factors affecting the development of their bodies and brains. Long-term or high-dose AAS treatment is likely to cause irreversible damage to their nervous system and psychological behavior, and these effects are easily overlooked. The article reviewed the long-term adverse effects of AAS on psychological behavior, emotion, cognitive functions and the nervous system of adolescents.
		                        		
		                        		
		                        		
		                        			Adolescent
		                        			;
		                        		
		                        			Anabolic Agents
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Cognition
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Nervous System
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Steroids
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Substance-Related Disorders
		                        			
		                        		
		                        	
7.Valproic acid withdrawal ameliorates impairments of hippocampal-spatial working memory and neurogenesis.
Wanassanun PANNANGRONG ; Apiwat SIRICHOAT ; Trai WONGSIRI ; Peter WIGMORE ; Jariya Umka WELBAT
Journal of Zhejiang University. Science. B 2019;20(3):253-263
		                        		
		                        			
		                        			Valproic acid (VPA), an agent that is used to treat epileptic seizures, can cause spatial memory impairment in adults and children. This effect is thought to be due to the ability of VPA to inhibit neurogenesis in the hippocampus, which is required for learning. We have previously used an animal model to show that VPA significantly impairs hippocampal-spatial working memory and inhibits neuronal generation in the sub-granular zone of the dentate gyrus. As there are patient reports of improvements in memory after discontinuing VPA treatment, the present study investigated the recovery of both spatial memory and hippocampal neurogenesis at two time points after withdrawal of VPA. Male Wistar rats were given intraperitoneal injections of 0.9% normal saline or VPA (300 mg/kg) twice a day for 10 d. At 1, 30, or 45 d after the drug treatment, the novel object location (NOL) test was used to examine spatial memory; hippocampal cell division was counted using Ki67 immunohistochemistry, and levels of brain-derived neurotrophic factor (BDNF) and Notch1 were measured using western immunoblotting. Spatial working memory was impaired 1 and 30 d after the final administration, but was restored to control levels by 45 d. Cell proliferation had increased to control levels at 30 and 45 d. Both markers of neurogenesis (BDNF and Notch1 levels) had returned to control levels at 45 d. These results demonstrate that memory recovery occurs over a period of six weeks after discontinuing VPA treatment and is preceded by a return of hippocampal neurogenesis to control levels.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Brain-Derived Neurotrophic Factor/metabolism*
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			Cognition/drug effects*
		                        			;
		                        		
		                        			Dentate Gyrus/drug effects*
		                        			;
		                        		
		                        			Enzyme Inhibitors/pharmacology*
		                        			;
		                        		
		                        			Hippocampus/metabolism*
		                        			;
		                        		
		                        			Immunohistochemistry
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Memory Disorders/therapy*
		                        			;
		                        		
		                        			Memory, Short-Term/drug effects*
		                        			;
		                        		
		                        			Neurogenesis/drug effects*
		                        			;
		                        		
		                        			Neurons/metabolism*
		                        			;
		                        		
		                        			Rats
		                        			;
		                        		
		                        			Rats, Wistar
		                        			;
		                        		
		                        			Receptor, Notch1/metabolism*
		                        			;
		                        		
		                        			Spatial Memory/drug effects*
		                        			;
		                        		
		                        			Valproic Acid/pharmacology*
		                        			
		                        		
		                        	
8.Gastrodin improves hippocampal neurogenesis by NO-cGMP-PKG signaling pathway in cerebral ischemic mice.
Huan XIAO ; Xiao-Jiao MA ; Ou-Mei CHENG ; Hong-Mei QIU ; Qing-Song JIANG
China Journal of Chinese Materia Medica 2019;44(24):5451-5456
		                        		
		                        			
		                        			This paper was aimed to investigate the effect of gastrodin( GAS) on hippocampal neurogenesis after cerebral was chemic and to explore its mechanism of action related to NO. The cerebral ischemia model of C57 BL/6 mice was established by bilateral common carotid artery occlusion. The pathological changes in hippocampal CA1 region and the cognitive function of mice were assessed by HE staining and Morris water maze test,respectively. The count of Brd U/Neu N positive cells in dentate gyrus was detected by immunofluorescence assay. The NOS activity and the NO content were determined by colorimetric and nitrate reduction methods,respectively.The level of c GMP was measured by ELISA kit,and the PKG protein expression was tested by Western blot. On postoperative day 8,the hippocampal CA1 pyramidal neurons of mice showed irregular structure,with obvious nuclear pyknosis,loose cell arrangement and obvious decrease in the number of neurons. On postoperative day 29,the spatial learning ability and memory were decreased. These results indicated cerebral ischemia in mice. Meanwhile,the Brd U/Neu N positive cells were increased significantly in ischemic mice,indicating that neurogenesis occurred in hippocampus after cerebral ischemia. Treatment with different doses of gastrodin( 50 and 100 mg·kg-1) significantly ameliorated the pathological damages in the CA1 region,improved the ability of learning and memory,and promoted hippocampal neurogenesis. At the same time,both the NOS activity and the NO concentration were decreased in model group,but the c GMP level was increased,and the PKG protein expression was up-regulated. Gastrodin administration activated the NOS activity,promoted NO production,further increased c GMP level and up-regulated PKG protein expression. These results suggested that gastrodin can promote hippocampal neurogenesis after cerebral ischemia and improve cognitive function in mice,which may be related to the activation of NO-cGMP-PKG signaling pathway.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Benzyl Alcohols/therapeutic use*
		                        			;
		                        		
		                        			Brain Ischemia/drug therapy*
		                        			;
		                        		
		                        			CA1 Region, Hippocampal/drug effects*
		                        			;
		                        		
		                        			Cognition
		                        			;
		                        		
		                        			Glucosides/therapeutic use*
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Mice, Inbred C57BL
		                        			;
		                        		
		                        			Neurogenesis
		                        			;
		                        		
		                        			Signal Transduction
		                        			
		                        		
		                        	
9.Effects of Honokiol on cognitive function in mice with kainic acid-induced epilepsy.
Qingmei WANG ; Min SHU ; Qianzi XU ; Yiyi XIE ; Shengzhe RUAN ; Jianda WANG ; Linghui ZENG
Journal of Zhejiang University. Medical sciences 2018;47(5):450-456
		                        		
		                        			OBJECTIVE:
		                        			To investigate the effects of Honokiol on cognitive function in mice with epilepsy.
		                        		
		                        			METHODS:
		                        			Kainic acid (38 mg/kg) was intraperitoneally injected in 5 weeks old male ICR mice to induce epilepsy. Honokiol at dose of 3, 10, 30 mg/kg was given to epilepic mice by intraperitoneal injection for 10 days. Fluoro-Jade B staining was used to assess neuronal death; Morris water maze and Y maze tests were used to measure cognitive function such as learning and memory; Western blot was performed to detect the expression of acetylated superoxide dismutase (SOD), microtubule associated protein 1 light chain 3-Ⅱ (LC3-Ⅱ) and P62 in hippocampus tissue; thiobarbituric acid and WST-1 methods were used to detect malondialdehyde (MDA) and SOD.
		                        		
		                        			RESULTS:
		                        			Compared with control group, the levels of acetylated-SOD, MDA, LC3-Ⅱ, P62 and neuronal death increased, cognitive function and SOD decreased in model group (<0.05 or <0.01). Honokiol at the dose of 10 mg/kg and 30 mg/kg decreased SOD acetylation, MDA content, expression of LC3-Ⅱ and P62, as well as neuronal death, and the cognitive function was improved (<0.05 or <0.01), especially in 30 mg/kg Honokiol group.
		                        		
		                        			CONCLUSIONS
		                        			Honokiol alleviates oxidative stress and autophagy degradation disorder, decreases neuronal death, and therefore improves cognitive function in epilepsy mice.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Biphenyl Compounds
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Cognition
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Epilepsy
		                        			;
		                        		
		                        			chemically induced
		                        			;
		                        		
		                        			Gene Expression Regulation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Hippocampus
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Kainic Acid
		                        			;
		                        		
		                        			Lignans
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Malondialdehyde
		                        			;
		                        		
		                        			Maze Learning
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Mice, Inbred ICR
		                        			;
		                        		
		                        			Neurons
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Superoxide Dismutase
		                        			;
		                        		
		                        			genetics
		                        			
		                        		
		                        	
10.Neuroprotective effects of Tongmai Yizhi Decoction () against Alzheimer's disease through attenuating cyclin-dependent kinase-5 expression.
Jing-Han FENG ; Bao-Chang CAI ; Wei-Feng GUO ; Ming-Yan WANG ; Yong MA ; Qiao-Xi LU
Chinese journal of integrative medicine 2017;23(2):132-137
OBJECTIVESTo explore the protective effects of Tongmai Yizhi Decoction (, TYD), a Chinese herb complex prescription against the impairment of cognitive functions and memory loss in amyloid beta 1-40 (Aβ) peptide and ibotenic (IBO)-induced Alzheimer's disease (AD) model rats.
METHODSThe in vivo model was established by injecting Aβand IBO into left hippocampal CA1 area of Sprague-Dawley (SD) rat to mimic AD. Totally 32 SD rats were divided into 4 groups, including sham operation group, AD model group, TYD group [AD rats treated with TYD at the dosage of 19.44 g/(kg•d) for 4 weeks] and huperzine A group [AD rats treated with huperzine A at the dosage of 40.5 μg/(kg•d) for 4 weeks]. Spatial learning and memory level was detected by Morris Water Maze test. Histological morphology in the hippocampus was tested by hematoxylin-eosin (HE) staining. Cyclin-dependent kinase-5 (Cdk5) protein and gene expression level were investigated by Western blot analysis and real-time quantitative polymerase chain reaction (RT-qPCR), respectively.
RESULTSAβ1-40 and IBO treatment induced longer escape latency of rats, compared with sham operation group from day 25 (P<0.01). However, TYD and huperzine A obviously shortened the escape latency from day 26 (P<0.01). Moreover, the effect of TYD was similar to huperzine A (P>0.05). Furthermore, HE staining also showed that TYD and huperzine A reversed the neuropathological changes in the hippocampus triggered by Aβ1-40 and IBO. TYD and huperzine A effectively reduced the expression levels of Cdk5 protein and gene located in rat hippocampus, compared with the AD model group (P<0.01).
CONCLUSIONTYD could be a promising neuroprotective agent for protecting neuron from AD injury through inhibiting Cdk5 expression.
Alzheimer Disease ; drug therapy ; pathology ; Animals ; Cognition ; drug effects ; Cyclin-Dependent Kinase 5 ; metabolism ; Disease Models, Animal ; Down-Regulation ; drug effects ; Drugs, Chinese Herbal ; therapeutic use ; Female ; Hippocampus ; drug effects ; Male ; Maze Learning ; drug effects ; Memory ; drug effects ; Neuroprotective Agents ; therapeutic use ; Rats ; Rats, Sprague-Dawley
            
Result Analysis
Print
Save
E-mail