文档库 最新最全的文档下载
当前位置:文档库 › 安颤灵治疗乳胞素诱导的帕金森病:与泛素-蛋白酶体通路有关

安颤灵治疗乳胞素诱导的帕金森病:与泛素-蛋白酶体通路有关

NEURAL REGENERATION RESEARCH Volume 7, Issue 3, January 2012

Cite this article as: Neural Regen Res. 2012;7(3):165-170.

165

Yinghong Li ☆, Doctor, Associate professor,

Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen 518035, Guangdong Province, China

Corresponding author: Zhengzhi Wu, Professor, Investigator, Doctoral supervisor, Shenzhen

Secon d People’s Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen 518035,

Guangdong Province, China; Shenzhen Institute of

Gerontology, Second Clinical Medical College, Jinan University, Shenzhen 518020, Guangdong Province, China

zhengzhw@https://www.wendangku.net/doc/8a13557695.html,

Received: 2011-06-02 Accepted: 2011-12-20 (N20101004002/YJ)

Li YH, Wu ZZ, Gao XW, Zhu QW, Jin Y , Wu AM, Huang CJA. Anchanling reduces pathology in a

lactacystin-induced

Parkinson's disease model. Neural Regen Res. 2012;7(3):165-170.

https://www.wendangku.net/doc/8a13557695.html,

https://www.wendangku.net/doc/8a13557695.html,

doi:10.3969/j.issn.1673-5374.2012.03.001

Anchanling reduces pathology in a lactacystin- induced Parkinson's disease model*☆○

Yinghong Li 1, Zhengzhi Wu 1, 2, Xiaowei Gao 1, Qingwei Zhu 1, Yu Jin 1, Anmin Wu 1, Andrew C. J. Huang 3

1Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzh en University, Shenzhen 518035, Guangdong Province, China 2Shenzhen Institute of Gerontology, Second Clinical Medical College, Jinan University, Shenzhen 518020, Guangdong Province, China 3Larry Hillblom Islet Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA 90024, USA

Abstract

A rat model of Parkinson's disease was induced by injecting lactacystin stereotaxically into the left mesencephalic ventral tegmental area and substantia nigra pars compacta. After rats were

intragastrically perfused with Anchanling , a Chinese medicine, mainly composed of magnolol, for 5 weeks, when compared with Parkinson’s disease model rats, tyrosine hydroxylase expression was increased, α-synuclein and ubiquitin expression was decreased, substantia nigra cell apoptosis was reduced, and apomorphine-induced rotational behavior was improved. Results suggested that Anchanling can ameliorate Parkinson's disease pathology possibly by enhancing degradation activity of the ubiquitin-proteasome system.

Key Words: Parkinson's disease; Anchanling ; ubiquitin-proteasome system; α-synuclein; tyrosine hydroxylase; cell apoptosis

INTRODUCTION

Loss of substantia nigra dopaminergic neurons and intracytoplasmic Lewy body formation are the main pathological changes in Parkinson's disease (PD)[1-4]. Lewy body formation is highly correlated with functional impairment of the ubiquitin-proteasome system (UPS)[5-9]. The UPS is an important pathway involving protein degradation in vivo , responsible for clearing mutated, damaged and misfolded proteins, and comprises

ubiquitin, ubiquitin-related enzymes and the proteasome. Abnormal changes in ubiquitin or ubiquitin-related enzymes E1, E2, E3 or protease may influence UPS function [10-12]. Recent evidence indicates that intracellular α-synuclein protein is misfolded or

overexpressed in response to α-synuclein gene mutations, which inhibits proteasome degradation and induces abnormal α-synuclein accumulation, resulting in neurotoxicity [13]. Overexpression of α-synuclein in nerve cells can induce

ubiquitin-dependent proteasome dysfunction and cell apoptosis [14]. Furthermore, proteasome dysfunction can result in

abnormal protein aggregation. Stereotaxic injection of the proteasome inhibitor

lactacystin into the substantia nigra is toxic to dopaminergic neurons, and induces protein aggregation and inclusion body

formation [15-16]. An in vitro experiment further demonstrated that lactacystin can induce

ubiquitinated protein aggregation and

promote intracytoplasmic ubiquitin-positive inclusion body formation [17].

Substitution therapy with levodopa has been commonly used to treat PD. It ameliorates PD symptoms, but induces severe toxicity and side effects and does not control PD progression. Recent studies demonstrated that Chinese medicines can delay PD

progression, relieve adverse effects due to Western medicine, and control symptoms of non-motor disturbance in PD [5-18]. However, studies mainly focused on oxidative stress, cell apoptosis, excitatory amino acid toxicity and immune reactions to investigate the mechanism of Chinese medicines [19-35]. Anchanling (ACL), a Chinese medicine, is effective in treating early stage PD, and exerts positive effects in PD patients with advanced stage disease, or poor response to dopamine therapy. However, the pharmacodynamic mechanism remains poorly understood. In the present study, a rat model of PD was established using lactacystin, and the effects of ACL on cell apoptosis and UPS function were observed by immunohistochemistry and terminal

deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) techniques.

RESULTS

Quantitative analysis of experimental animals

Following one-week adaptation, 60 of 80

https://www.wendangku.net/doc/8a13557695.html,

Sprague-Dawley rats were selected according to their food intake, behavior and coat[36-37]. Lactacystin was stereotaxically injected in the left mesencephalic ventral tegmental area (VTA) and substantia nigra pars compacta (SNc) of 50 rats to establish a PD model. A total of 49 rats survived after lesion, and 27 were selected following screening of apomorphine-induced behavior. Ten were used as the vehicle-treated (PD) group and 10 as the ACL group, respectively treated with distilled water and ACL by intragastric perfusion. Another 10 of 60 rats were used as the control group. Therefore, 30 rats were included in the final analysis.

Influence of ACL on substantia nigra tyrosine hydroxylase (TH) expression in lesioned rats Immunofluorescent labeling showed that TH was expressed in cells of the substantia nigra of control group rats and the number of TH-positive cells was 293.8 ±13.0 per field of view (× 200) (n = 6). After 5 weeks on vehicle treatment, the number of TH-positive cells was significantly reduced in the lesioned group (53.50 ± 14.05 per field of view (× 200); n = 6) compared with control group (P < 0.05). 5 weeks of ACL increased TH-positive cells in the substantia nigra of rats (130.33 ± 11.91 per field of view (× 200); n = 6) compared with the

vehicle-treated group (P < 0.05), but this remained lower than the control group (P < 0.05; Figure 1).

Influence of ACL on substantia nigra α-synuclein and ubiquitin expression in lesioned rats Immunofluorescence and thioflavin S (a chromogenic marker of amyloid substance) labeling were used to examine protein aggregation. The rate of thioflavin S and α-synuclein double labeling, as well as thioflavin S and ubiquitin double labeling was significantly increased at 5 weeks in the vehicle-treated group compared with the control group. ACL significantly reduced thioflavin S and α-synuclein double labeling, as well as thioflavin S and ubiquitin double labeling (Figures 2, 3).

Figure 2 Alpha-synuclein protein expression in rat substantia nigra (immunofluorescence double-labeling staining, × 200). Thioflavin S was used as a chromogenic marker of amyloid substance. Red fluorescence represents α-synuclein staining, and green represents thioflavin S staining. In the vehicle-treated (model) group, co-labeling of α-synuclein and thioflavin S was

evident. In the Anchanling group, the co-labeling was reduced compared with the model group. Only weak red and green fluorescence was observed in the control group.

A

n

c

h

a

n

l

i

n

g

V

e

h

i

c

l

e

-

t

r

e

a

t

e

d

C

o

n

t

r

o

l

Cy3-labeled α-synuclein staining Thioflavin S staining Co-labeling

Figure 1 Substantia nigra tyrosine hydroxylase (TH)

expression in rats (immunofluorescent staining, × 200).

TH-positive reaction was represented by red fluorescence.

A large number of TH-positive cells were observed in the

control group (A). Compared with the control group, the

number of TH-positive cells was significantly decreased in

the vehicle-treated (model) group (B). Anchanling (C)

increased TH-positive cells in rats compared with the

vehicle-treated group.

A

B

C

166

Influence of ACL on nigral apoptosis in lesioned rats

TUNEL showed that cell apoptosis was significantly

increased in the substantia nigra of the vehicle treated

group compared with the control group (P < 0.05). However,

it was significantly decreased in the ACL group compared

with the vehicle treated group (P < 0.05; Table 1).

Influence of ACL on behavior in lesioned rats

Three weeks after lesion locomotion was significantly

reduced. Crawling movements and escape responses to

external stimuli were delayed. Spontaneous rotational

behavior, sniffing and back bending appeared. At five

weeks, all rats underwent apomorphine induction.

Rotation toward the healthy side was significantly

increased in the vehicle-treated group and ACL group

(P < 0.05), but the frequency of apomorphine-induced

rotation in the Anchanling group is less than

vehicle-treated group (P < 0.01). The control group rats

did not show aberrant rotational behavior (Table 2).

DISCUSSION

In the present study, PD symptoms were induced in rats

by injection of lactacystin into the SNc and VTA. As

previously published[38], thioflavin S was used as a

chromogenic marker of amyloid substance to observe

abnormal protein aggregation in cells.

Immunofluorescent double labeling showed stronger

co-staining of α-synuclein and thioflavin S in the lesioned,

vehicle-treated group compared with the control group,

suggesting α-synuclein aggregation in the former. In

addition, co-staining of ubiquitin and thioflavin S was

evident in the vehicle-treated group, indicating

ubiquitination of amyloid protein. TUNEL showed that

cell apoptosis in the substantia nigra was significantly

increased in the vehicle-treated group compared with the

control group. Therefore, microinjection of lactacystin

into the SNc and VTA can successfully induce

Figure 3 Ubiquitin protein expression in rat substantia nigra (immunofluorescence double-labeling staining, × 200).

Thioflavin S was used as chromogenic marker of amyloid substance. Red fluorescence represents ubiquitin staining, and green represents thioflavin S staining. In the vehicle-treated (model) group, co-labeling of ubiquitin and thioflavin S was evident. In the Anchanling group, the co-labeling was reduced compared with the model group. Only weak red and green fluorescence was observed in the control group.

A

n

c

h

a

n

l

i

n

g

V

e

h

i

c

l

e

-

t

r

e

a

t

e

d

C

o

n

t

r

o

l

Cy3-labeled α-synuclein staining Thioflavin S staining Co-labeling

167

pathological characteristics of PD. This is consistent with a correlation between proteasome dysfunction,

α-synuclein aggregation and cell apoptosis in the substantia nigra. That is, functional disturbance of proteasomes can induce α-synuclein aggregation and cell apoptosis in the substantia nigra. Proteasome dysfunction can trigger cell cycle signals in dopaminergic neurons by upregulating cell cycle regulating factor expression, thereby inducing neuronal apoptosis and degeneration[36]. The proteasomal inhibitor lactacystin can activate caspase-3 and induce PC12 apoptosis[37]. Therefore, we conclude that lactacystin inhibits proteasome function in the UPS, thereby blocking

α-synuclein degradation, inducing α-synuclein aggregation, cell structural damage, triggering apoptosis, and leading to cell degeneration and death.

In the present study, ACL reduced α-synuclein aggregation and cell apoptosis in the substantia nigra compared with the vehicle-treated group, possibly by improving proteasomal degradation of α-synuclein, reducing protein aggregation and inclusion body formation, and/or activating apoptosis-inhibiting signal pathways to protect nigral cells.

In conclusion, UPS dysfunction can induce α-synuclein and ubiquitin aggregation and inclusion body formation, resulting in neuronal apoptosis in the substantia nigra. ACL may ameliorate PD pathology by enhancing the function of the ubiquitin-proteasome system. MATERIALS AND METHODS

Design

A randomized, controlled, animal experiment was used. Time and setting

The experiment was performed at the SPF Animal Laboratory, Shenzhen Institute for Biochemical Drug Control, and Central Laboratory of First Affiliated Hospital of Shenzhen University, China from June 2007 to February 2009.

Materials

Animals

A total of 80 healthy, adult, male Sprague Dawley rats, weighing 200-250 g, were provided by the Medical Animal Experimental Center of Guangdong Province (No. SCXK (Yue) 2003-0002; 2006A015). The rats were housed in humidity of 40-70% at 20-26°C. The experimental procedures were performed in accordance with the Guidance Suggestions for the Care and Use of Laboratory Animals, issued by the Ministry of Science and Technology of the Peop le’s Republic of China[39]. Drugs

ACL was provided by the Shenzhen Institute of Gerontology. The crude drugs were purchased from Shenzhen Accord Pharmaceutical Co., Ltd., China. ACL comprised magnolia bark, fourstamen stephania root and Chinese Magnolivine Fruit and prepared by routine water-ethanol method[40]. Suspension was prepared by mixing with distilled water and administrated intragastrically. The active component was magnolol (5, 5'-Diallyl-2, 2'-biphenyldiol, molecular weight 266.32) and honokiol (3’, 5-Diallyl-2-4’-dihydroxybiphenyl, molecular weight 266.33). They were isomeric[41]. Their chemical structural formulas are shown in Figure 4. Methods

Establishment of PD model

Following behavioral tests, animals without rotational behavior were included for subsequent experiments. The rats were anesthetized by intraperitoneal injection of 2% pentobarbital (45 mg/kg; Sigma, St. Louis, MO, USA), and placed at the stereotaxis instrument (RWD Life Science, Shenzhen, China). The head was routinely sterilized, and the scalp was cut open, and the periosteum was dissected. According to rat stereotaxic atlas (supplementary Figure 1 online), SNc and VTA location was identified[42]: SNc: 5.0 mm posterior to bregma, 1.7 mm left to stereotaxic atlas, 7.6 mm below cranium surface; VTA: 4.6 mm posterior to bregma,

0.9 mm left to stereotaxic atlas, 7.5 mm below cranium surface. According to body mass and cranium size, the coordinates were confirmed and marked using three edged needle. The cranium was drilled using dental drill, and a microsyringe (Hamilton, CH-7402 Bonaduz, GR, Switzerland) was slowly inserted according to coordinates to inject lactacystin (Sigma; 8 μg (4μL) per point) in vehicle-treated and ACL groups, while the control group was injected with the same volume of normal saline. The injection was inserted to a depth of the center of pinhole slope (zero at cerebral dura mater), 1 μL/min. The needle was maintained for 10 minutes, and slowly removed. The wound was sutured, followed by intraperitoneal injection of penicillin (10 × 104 U/kg/d) for one week to prevent infection. In addition, the wound was disinfected with iodine tincture, once a day. The sutures were removed seven days later.

Evaluation of model

Behaviors were observed at 9: 00 am every seven days from one week postoperatively. The rats were intraperitoneally injected with 0.5 mg/kg apomorphine (Sigma). The rotational behaviors of rats were observed in a plastic tub (40 cm diameter) and rotations within five minutes after injection were recorded for three weeks. Successful lesioning was identified if the rats rotated towards the right within 30 minutes, with mean speed > 7 r/min. Rats with rotations towards the left or rotations towards the right at < 7 r/min or without rotation were excluded.

Figure 4 Chemical structures of honokiol (A) and

magnolol (B).

A B

168

ACL administration

The ACL group rats were administrated ACL at a dose of 10 times of that for a 60 kg human, dissolved in normal saline, 1.5 mL/100 g[43], 9: 00-10: 00 a.m., once a day. Animals were weighed every week, and the dose was adjusted accordingly. The drugs were administrated for five consecutive weeks. The vehicle-treated and control groups were perfused with the same volume of normal saline. All rats were administrated immediately following model establishment.

Behavioral detection

The rats were induced with apomorphine at 1, 3, 5 weeks after ACL administration to observe rotations and other abnormal behaviors. Changes in rotation frequency before and after administration and among groups were compared.

Immunofluorescent staining for TH expression in the substantia nigra

Five weeks after administration, two rats were randomly selected from each group, anesthetized by intraperitoneal injection with 2% pentobarbital (45 mg/kg), followed by perfusion of 4% paraformaldehyde via left ventricular intubation. Brains were harvested, fixed in postfixation solution containing 20% sucrose at 4°C for 2 hours, followed by postfixation solution containing 30% sucrose at 4°C for 2 hours. Serial coronal sections were prepared originating from the substantia nigra using freezing microtome (Microm HN525, Therm, Walldrorf, Germany), 30 μm thick. Symmetrical sections from the substantia nigra were selected for TH immunohistochemistry. The sections were washed with 0.01 M phosphate buffered saline (PBS) (pH 7.4), blocked in 2% bovine serum albumin at room temperature for 30 minutes, incubated with mouse anti-TH polyclonal antibody (1: 200; Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA) at 4°C overnight, washed, incubated with Cy3-labeled goat

anti-rat secondary antibody dilution (1: 200; Santa Cruz Biotechnology Inc.) at 37°C for 1 hour, shaken, rinsed, mounted with 50% neutral resin, and observed by fluorescence microscope (Leica Inc.). The mean number of TH-positive cells in bilateral substantia nigra sections under 200 × magnification was calculated. Immunofluorescent double labeling for α-synuclein and ubiquitin in the substantia nigra

The sections were washed with 0.01 M PBS (pH 7.4), blocked in 2% bovine serum albumin at room temperature for 30 minutes, incubated with mouse anti- α-synuclein (1: 500) and ubiquitin polyclonal antibodies (1: 400; Sigma) at 4°C overnight, washed, incubated with Cy3-labeled goat anti-mouse secondary antibody dilution (1: 200; Santa Cruz Biotechnology Inc.) at 37°C for 1 hour, followed by 0.1% thioflavin S (Sigma) for 10 minutes. The sections were differentiated with 80% ethanol for 5 minutes, shaken, rinsed, mounted with 50% neutral resin, and observed by fluorescence microscope. TUNEL detection for cell apoptosis in the substantia nigra

After administration, two rats were randomly selected from each group, anesthetized, and the midbrain was harvested, fixed in 4% paraformaldehyde for 24 hours, dehydrated by gradient ethanol, paraffin embedded, followed by coronal sectioning, 5 μm thick. Five sections were selected from each block of brain tissues. TUNEL was performed according to TUNEL kit (R&D Systems, Minneapolis, MN, USA). Briefly, the sections were dewaxed, and washed with PBS, 3 × 5 minutes after antigen was retrieved by microwave, incubated with 20% normal goat serum at room temperature for 30 minutes. TUNEL reaction solution was added to the sections and incubated at 37°C for 90 minutes, washed with PBS, 3 × 5 minutes, followed by 3% H2O2 methanol solution at room temperature for 10 minutes and incubation at 37°C for 90 minutes. The sections were treated with POD transforming agent at 37 °C for

30 minutes, washed with PBS, 3 × 5 minutes, visualized with diaminobenzidine/H2O2, stained with hematoxylin, dehydrated, cleared, and mounted with neutral resin. Six sections were selected from each group, and three 4 mm × 4 mm fields of view were selected. The number of TUNEL-positive cells and apoptotic index were analyzed using Leica Qwin image analysis system software (Leica Microsystems Imaging Solutions Ltd., Software Version 3.0, Cambridge, UK).

Statistical analysis

Data were analyzed using SPSS version 10.0 (SPSS, Chicago, IL, USA) and are expressed as mean ± SD. In homogeneity of variance, data were analyzed using repeated measurement of analysis of variance,

one-way analysis of variance or least significant difference-t test; while data were analyzed with Tamhane test in heterogeneity of variance.

Author contributions:Yinghong Li analyzed data, revised manuscript, provided technical and data support. Zhengzhi Wu conceived and designed this study, was in charge of funds, and revised manuscript. Xiaowei Gao provided experimental data and contributed to statistical analysis, and wrote manuscript. Qingwei Zhu conducted the experiments, provided technical and data support. Yu Jin, Anmin Wu and Andrew C. J. Huang provided technical and data support.

Conflicts of interest: None declared.

Funding: This study was supported by the National Natural Science Foundation of China (Influence of Anchanling on endoplasmic reticulum stress and ubiquitin-proteasome system in PD model), No. 30772757.

Ethical approval:This study received permission from the Animal Ethnics Committee of First Affiliated Hospital of Shenzhen University, China.

Acknowledgments: We thank the staff of Animal Laboratory, Shenzhen Institute for Biochemical Drug Control, and Clinical Institute of Chinese and Western Medicine, First Affiliated Hospital of Shenzhen University, China, for technical support. Supplementary information: Supplementary data associated with this article can be found, in the online version, by visiting https://www.wendangku.net/doc/8a13557695.html,, and entering Vol. 7, No. 3, 2012 after selecting the “NRR Current Issue” button on the page.

169

REFERENCES

[1] Dauer W, Przedborski S. Parkinson's disease: mechanisms and

models. Neuron. 2003;39(6):889-909.

[2] Giasson BI, Lee VM. Are ubiquitination pathways central to

Parkinson's disease? Cell. 2003;114(1):1-8.

[3] Greenamyre JT, Hastings TG. Biomedicine.

Parkinson's--divergent causes, convergent mechanisms. Science.

2004;304(5674):1120-1122.

[4] Lansbury PT Jr, Brice A. Genetics of Parkinson's disease and

biochemical studies of implicated gene products. Curr Opin Cell

Biol. 2002;14(5):653-660.

[5] Bukhatwa S, Zeng BY, Rose S, et al. A comparison of changes in

proteasomal subunit expression in the substantia nigra in

Parkinson's disease, multiple system atrophy and progressive

supranuclear palsy. Brain Res. 2010;1326:174-183.

[6] Lee FK, Wong AK, Lee YW, et al. The role of ubiquitin linkages on

alpha-synuclein induced-toxicity in a Drosophila model of

Parkinson's disease. J Neurochem. 2009;110(1):208-219.

[7] Olanow CW, McNaught KS. Ubiquitin-proteasome system and

Parkinson's disease. Mov Disord. 2006;21(11):1806-1823.

[8] Betarbet R, Sherer TB, Greenamyre JT. Ubiquitin-proteasome

system and Parkinson's diseases. Exp Neurol. 2005;191 Suppl 1: S17-27.

[9] Barrachina M, Casta?o E, Dalfó E, et al. Reduced ubiquitin

C-terminal hydrolase-1 expression levels in dementia with Lewy

bodies. Neurobiol Dis. 2006;22(2):265-273.

[10] Heo JM, Rutter J. Ubiquitin-dependent mitochondrial protein

degradation. Int J Biochem Cell Biol. 2011;43(10):1422-1426. [11] Taylor EB, Rutter J. Mitochondrial quality control by the ubiquitin-

proteasome system. Biochem Soc Trans. 2011;39(5):1509-1513.

[12] Egeler EL, Urner LM, Rakhit R, et al. Ligand-switchable

substrates for a ubiquitin-proteasome system. J Biol Chem. 2011;

286(36):31328-31336.

[13] Forloni G, Bertani I, Calella AM, et al. Alpha-synuclein and

Parkinson's disease: selective neurodegenerative effect of alpha- synuclein fragment on dopaminergic neurons in vitro and in vivo.

Ann Neurol. 2000;47(5):632-640.

[14] Stefanis L, Larsen KE, Rideout HJ, et al. Expression of A53T

mutant but not wild-type alpha-synuclein in PC12 cells induces

alterations of the ubiquitin-dependent degradation system, loss of dopamine release, and autophagic cell death. J Neurosci. 2001;

21(24):9549-9560.

[15] Zhao J, Liu ZG, Chen SD, et al. The degeneration of dopaminergic

neuron and inclusion-like formation induced by the inhibitor of

proteasome. Zhongfeng yu Shenjing Jibing Zazhi. 2004;21(6):

493-495.

[16] Zhang KZ, Wang J, Ding ZT, et al. The roles of proteasome on

nigral degeneration and Lewy body formation. Zhonghua Laonian Yixue Zazhi. 2004;23(4):259-262.

[17] Yang H, Chen SD, Li BZ, et al. The block of ubiquitin-proteasome

pathway induces cell death and the formation of ubiquitin-

immunoreactive inclusions in PC12 cells. Zhonghua Shenjing Ke

Zazhi. 2005;38(7):430-433.

[18] Niu C, Zhang J, Mei J, et al. Does neurotrophic factor benefit to

PD therapy via co-function with ubiquitin-proteasome system?

Med Hypotheses. 2011;76(4):589-592.

[19] Wang YC, Xu HL, Fu Q, et al. Resveratrol derived from Rhizoma

Et Radix Polygoni Cuspidati and its liposomal form protect nigral

cells of Parkinsonian rats. Zhongguo Zhongyao Zazhi. 2011;56(8): 1060-1066.

[20] Niu JY, Xu F. The protective effect and mechanism of green tea

polyphenols in substantia nigra dopaminergic neurons of

Parkinson's disease. Shandong Yiyao. 2010; 54(11):56-57. [21] He JC, Wang ZH, Yuan CX, et al. Effects of compound rehmannia

prescription on the neuroethology and oxidative stress in rats with Parkinson's disease. Zhongguo Kangfu Yixue Zazhi. 2009;25(7):

590-592. [22] Dong MX, Niu YC, Pan Z, et al. The protective effect of pueraria

isoflavones on MPP+-induced oxidative stress in the PC12.

Zhongyao Yaoli yu Linchuang. 2009;26(2):48-49.

[23] Gao LX, Liu K. Summary of Chinese medicine treatment of

Parkinson's disease. Shizhen Guoyi Guoyao. 2010;21(3):

724-725.

[24] Li HL, Shu Y, Hou RR, et al. Neuroprotective effects of gastrodin

on PQ and MB Induced dopaminergic neurons damage in C_(57)

BL mice. Chengdu Zhongyiyao Daxue Xuebao. 2010;53(1):57-59.

[25] Zhang L, Liu SM. Acanthopanax prevention Parkinson’s disease.

Zhongguo Zhongyiyao Xinxi Zazhi. 2007;17(3):95-96.

[26] Yuan HL, Wang X, Zhang LP, et al. Mechanism and research

progress of Chinese traditional medicine in the prevention and

treatment of Parkinson's disease. Zhongguo Yaoli Xue Tongbao.

2010;26(7):850-854.

[27] Dong MJ, Qian HY, Zhou SF, et al. Effect of Liuwei Rehmanniae

Pill on Mouse's Oxidation Stress Reaction with Parkinson.

Zhejiang Zhongyiyao Daxue Xuebao. 2009;34(6):756-757. [28] Ding HJ, He JC, Wang WW. Effects of Ditan decoction on

behavioristics and oxidative stress reaction of rats with

parkinson's disease. Shanghai Zhongyiyao Zazhi. 2009;56(3):

63-65.

[29] Wang WW, He JC, Ding HJ. Effect of tianma gouteng drink on the

behavioural and oxidation stress response of Parkinson’s disease rat. Zhongguo Laonian Xue Zazhi. 2010;30(12):1657-1659. [30] Guo SH, Bezard E, Zhao BL. Protective effect of green tea

polyphenols against 6-OHDA induced apoptosis in SH-SY5Y cells through ROS-NO pathway. Bopu Xue Zazhi. 2006;28(4):550-551.

[31] An LJ, Li ZG, Jiang B. Parkinson's disease and cell apoptosis.

Zhongguo Xiandai Yixue Zazhi. 2006;20(7):1032-1036.

[32] Chen JZ, Huang C, Li XM, et al. Research clew and practice of

treating Parkinsonism with method of tonifying liver and kidney.

Zhongguo Yiyao Xuebao. 2004;19(11):688.

[33] He JC, Yuan SX, Wei HC, et al. The effect of nourishing the liver

and kidney meridians and detoxifying herbs on cell apoptosis in

PD rats. Zhonguo Laonian Xue Zazhi. 2003;22(5):217.

[34] Zhu WF, Luo RJ, Zhou LP, et al. Experimental studies on

Zhenchan Ning for Parkinson's disease. Guangzhou Zhongyiyao

Daxue Xuebao. 2004;21(4):284.

[35] Cai DF, Chen XQ, Gao Y, et al. Effect of Bushen Yanggan recipe

on nigrostriatal function in Parkinsonian model rats after long-term levodopa treatment. Zhongguo Zhongxiyi Jiehe Zazhi. 2002;22(1):

43.

[36] Zhang ZT, Cao XB, SunSG, et al. The effect and mechanism of

proteasome inhibitor induced cell cycle reentry on dopaminergic

neurons. Zuzhong yu Shenjing Jibing. 2006;13(3):134-137. [37] Yang H, Chen SD, Lu GQ, et al. Proteasomal inhibitor lactacystin

induces cell apoptosis and caspase 3 activation in PC12 cells.

Zhonghua Yixue Zazhi. 2005;85(29):2058-2061.

[38] Gao XW. Anchang ling’s effection on ubiquitin prosbasome system

of parkinson’s disease rat models. Wuhan: Hubei University of

Traditional Chinese Medicine. 2007.

[39] The Ministry of Science and Technology of the People’s Republic

of China. Guidance Suggestions for the Care and Use of

Laboratory Animals. 2006-09-30.

[40] Guo Y, Liang XY, Nian WL. Preliminary studies on extracting

aqueous depsides from Salvia Yunanensis with water-alchohol

methods. Yunnan Zhongyi Xueyuan Xuebao. 2001;24(4):6-8. [41] Sun AL, Feng L, Liu RM. Preparative isolation and purification of

honokiol and magnolol from Magnolia Officinali s Rehd. et Wils by high-speed countercurrent chromatography. Fenxi Huaxue. 2005;

33(7):1016-1018.

[42] Bao XM, Shu SY. The Rat Brain in Stereotaxic Coordinates.

Beijing: People’s Medical Press. 1991.

[43] Shi XQ. Experiments Methods of Medical Animals. Beijing:

People’s Medical Press. 1986.

(Edited by Zhao Y, Wang Z/Su LL/Wang L)

170

相关文档
相关文档 最新文档