Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Okajimas Folia Anat. Jpn.

, 83(1): 1–6, May, 2006

Effects of Chronic Ethanol Administration on the Expression


Levels of Neurotrophic Factors in the Rat Hippocampus
By

Hanayo OKAMOTO1, Takanori MIKI2, Kyoung-Youl LEE2, Toshifumi


YOKOYAMA2, Hiromi KUMA1, He GU2, Hong-Peng LI2, Yoshiki
MATSUMOTO2, Ippei YAMAOKA2, Kazutoshi FUSUMADA2, Tomohiro
IMAGAWA3, Zhi-Yu WANG2, Yu NAKAMURA1 and Yoshiki TAKEUCHI2

1Department of Neuropsychiatry, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan


2Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan
3Department of Veterinary Anatomy, Faculty of Agriculture, Tottori University, Tottori 680-8553, Japan

– Received for Publication, December 21, 2005 –

Key Words: Ethanol, Hippocampus, BDNF, GDNF, Real-time PCR

Summary: Chronic ethanol consumption has adverse effects on the central nervous system. Hippocampus is one of the
target sites of ethanol neurotoxicity. Hippocampal damage is known to result in impairment of learning and memory.
This study was aimed to determine whether chronic ethanol consumption could alter the expression levels of brain-
derived neurotrophic factor (BDNF) and glial-derived neurotrophic factor (GDNF) mRNAs in the hippocampus. Male
Wistar rats were given unrestricted access to a liquid diet containing 5% (v/v) ethanol as the sole fluid source for 19
weeks beginning at 10 weeks of age. The expression levels of BDNF and GDNF mRNAs in the hippocampus were
analyzed by real-time reverse transcription-polymerase chain reaction (RT-PCR) analysis. The present study revealed
that chronic ethanol consumption did not result in significant changes in the expression levels of BDNF and GDNF
mRNAs. Our present results showed no significant alteration in the expression of these neurotrophic factors; these re-
sults will lead to further studies to examine the possible alterations in the gene expression of various neurotrophins that
are related to hippocampal functions including learning and memory.

Chronic ethanol ingestion has adverse effects on the hippocampus showed altered immunoreactiv-
the central nervous system (CNS). In fact, numer- ities to calbindin-D28k and glial fibrillary acidic
ous studies on laboratory animals and humans have protein (GFAP), leading to altered neuron-glial in-
demonstrated that long-term ethanol consumption teractions4). This has led to the speculation that the
can result in impairment of cognition, learning, and altered immunoreactivities in the hippocampus are
memory1–3). It is now well known that even un- associated with various neurological symptoms seen
complicated alcoholics without specific neurological in alcoholic patients, such as cognitive dysfunction
or hepatic problems exhibit signs of regional brain and disabilities in orientation or learning. We have
damage and cognitive dysfunction. The hippo- now extended this study to evaluate these alter-
campus, which is known as one of the target sites ations in the hippocampus with respect to changes
for neurotoxic effects, is more sensitive than other in the expression levels of mRNAs of various
brain regions and plays an important role in learn- neurotrophic factors.
ing and memory processing. Brain-derived neurotrophic factor (BDNF) and
We have recently reported that in mice that glial-derived neurotrophic factor (GDNF) are
showed intoxication signs following ethanol treat- members of the neurotrophin family of neuro-
ment for several days, the neurons and astrocytes in trophic factors that are abundantly and widely dis-

* Corresponding author: Takanori Miki, Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, 1750-1
Ikenobe, Miki-cho, Kagawa 761-0793, Japan. E-mail address: mikit@med.kagawa-u.ac.jp

1
2 H. Okamoto et al.

tributed in the hippocampus5,6). BDNF is involved 60 min, followed by 65 C for 10 min using Ready-
in the survival of neurons and maintenance of neu- To-Go You-Prime First-Strand Beads (Amersham
ronal functions and participates in neuroprotection Biosciences, Piscataway, USA). For a 33-mL reac-
against various insults. GDNF is a member of the tion mixture, the following reagents were used:
transforming growth factor superfamily and is dis- 1 mL of sample RNA, 29 mL of RNase-free water,
tributed throughout multiple brain regions; it also Ready-To-Go You-Prime First-Strand Beads, and
has neuroprotective functions7–9). 3 mL of oligo dT primer (10 mM). Real-time PCR
Various insults such as alcoholic, epileptic, is- was performed using a LightCycler rapid thermal
chemic, and traumatic insults can induce marked cycler system (Roche Diagnostics Ltd, Lewes, UK).
changes in the level of gene expression of neuro- The reactions were performed in a 20-mL volume
trophins, ultimately leading to functional alter- with 2 mL of the cDNA diluted ten times, 0.5 mM
ations in the CNS10). It is of interest to focus upon primers, and reagents included in the LightCycler-
BDNF and GDNF separately since they have been FastStart DNA Master SYBR Green I mix (Roche
known to possess different signal transduction sys- Diagnostics GmbH, Mannheim, Germany). The
tems7). The purpose of the present study was to as- amplification protocol consisted of one cycle at
certain whether chronic ethanol administration may 95 C for 10 min, followed by 30 cycles at 95 C for
alter the level of gene expression of the two differ- 10 s, 65 C for 10 s, 72 C for 20 s, and 87 C for 2 s.
ent neurotrophic factors, i.e., BDNF and GDNF. Detection of the fluorescent products was carried
out at the end of the 87 C extension period. Re-
garding GDNF, the temperature for annealing
Materials and Methods was set at 58 C. To assess an appropriate inter-
nal control, coamplification of the housekeeping
Male Wistar rats obtained from CLEA Japan gene glyceraldehyde-3-phosphate dehydrogenase
(Tokyo, Japan) were used in the present study. (GAPDH) was performed in each sample. The fol-
The rats were divided into two groups, ethanol- lowing forward (F) and reverse (R) primers were
fed and pair-fed control. The experimental rats used in the present study: BDNF (gene accession
(n ¼ 6) were given unrestricted access to a liquid number, X67108), F: GAT GAG GAC CAG AAG
diet (Oriental yeast, Tokyo, Japan) containing 5% GTT CG, R: GAT TGG GTA GTT CGG CAT
(v/v) ethanol (99.5%; Wako, Osaka, Japan) as the TG; GDNF (NM019139), F: CCC GAA GAT TAT
sole fluid source for 19 weeks (beginning at 10 CCT GAC CA, R: TAG CCC AAA CCC AAG
weeks of age). The pair-fed control rats (n ¼ 6) TCA GT; GAPDH (AB017801), F: GTA TTG
were fed an identical liquid diet, except that sucrose GGC GCC TGG TCA CC, R: CGC TCC TGG
was substituted isocalorically for ethanol. This AAG ATG GTG ATG G. The above series of real-
study was conducted in compliance with the guide- time PCR procedure was performed three times.
lines for experimental use and care of laboratory Melting curve analysis and electrophoresis were
animals established by the Kagawa University Ani- performed to confirm amplification specificity and
mal Ethics Committee. exclude genomic contamination, respectively, as
At 29 weeks of age, the rats in both groups were described previously11). For the melting curve
anesthetized with sodium pentobarbital (60 mg/kg, analysis, the PCR products were melted by gradu-
i.p.) and perfused intracardially with medical-grade ally increasing the temperature beginning at 65 C
physiological saline (Otsuka, Tokyo, Japan). Using in 0.2 C steps. Electrophoresis of the PCR products
a vibratome, the brains were sectioned in the hori- amplified from cDNA using BDNF, GDNF, and
zontal plane to yield 1-mm-thick slices. The hippo- GAPDH primers was carried out on a 2% agarose
campal region was removed in chilled physiological gel, and the gel was then stained with ethidium
saline by using a dissecting microscope. Total RNA bromide. A similar electrophoresis of the amplifi-
was extracted by homogenizing these hippocampal cation products without RT was also performed for
slices in TRIzol reagent (Invitrogen, Carlsbad, each sample as a negative control.
USA). The concentration and purity of the ex- Statistical analysis of the body and brain weights
tracted RNA were evaluated by optical density was performed using Mann-Whitney U test or Stu-
measurements at 260 nm and 280 nm using a spec- dent’s t-test. The quantification data for mRNA
trophotometer. The RNA samples were stored at was analyzed using the LightCycler analysis soft-
80 C until use. ware. Data on the mRNA of BDNF and GDNF
Real-time reverse transcription-polymerase were expressed as the ratio of the expression levels
chain reaction (Real-time RT-PCR) analysis was of BDNF or GDNF mRNA to that of GAPDH
performed as described previously11). In brief, re- mRNA. Statistical analysis with Mann-Whitney
verse transcription was carried out at 37 C for U test or Student’s t-test was performed by using
Chronic Ethanol Administration and Neurotrophic Factors 3

Table 1. Data on the body weights (g) of chronic ethanol-fed SigmaStat software (Systat Software, Point Rich-
and pair-fed control rats at 10 and 29 weeks of age mond, USA). When the data was not normally dis-
tributed and the variances between groups were not
Group n 10 weeks of age 29 weeks of age
homogeneous, the square root transformation of
Chronic ethanol-fed 6 295.0 G 1.8 393.3 G 20.9* the data was performed prior to statistical analysis.
Pair-fed control 6 294.2 G 2.0 527.5 G 3.6

Each value is represented as mean G SEM.


Results
n: number of animals examined.
The body weight data were analyzed using Mann-Whitney U test
because the variances were not homogeneous. The mean G SEM values of the body weights of
* p < 0.01 chronic ethanol-fed and pair-fed control rats are
shown in Table 1. At the beginning of ethanol
treatment (10 weeks of age), no significant differ-
ence in body weight was observed between the two
Table 2. Data on the brain weights (g) of chronic ethanol-fed groups; however, at the end of 19 weeks of ethanol
and pair-fed control rats at 29 weeks of age
treatment, a significant difference was observed.
Group n Brain weights (g) Table 2 shows the mean G SEM values of the brain
weights of chronic ethanol-fed and pair-fed control
Chronic ethanol-fed 6 2.05 G 0.01* rats at the end of ethanol treatment (29 weeks of
Pair-fed control 6 2.23 G 0.02 age); statistical analysis using Mann-Whitney U test
Each value is represented as mean G SEM.
revealed a significant difference between their brain
n: number of animals examined. weights.
* p < 0.01 Figure 1 shows a representative electrophoresis

Fig. 1. A representative agarose gel electrophoresis of the PCR products amplified from cDNA using BDNF, GDNF, and GAPDH
primers; the electrophoresis shows a single band. No band was observed when a similar amplification procedure was per-
formed without RT (data not shown). A similar electrophoresis of the PCR products was carried out for each sample in order
to exclude genomic contamination. Molecular weight markers are shown in the left lane.
4 H. Okamoto et al.

Despite numerous studies on the effects of


Table 3. Data on the ratio (10 2 ) of the expression levels of chronic ethanol treatment on various neurotrophic
BDNF and GDNF mRNAs to that of GAPDH factors in the CNS, consistent results have not been
mRNA in chronic ethanol-fed and age-matched pair-
fed control rats
obtained. The differences in results across labo-
ratories may arise due to variations in experimental
Group n BDNF GDNF designs such as the length of ethanol treatment,
ethanol concentration in the diet, sampling time
Chronic ethanol-fed 6 33:1 G 0:7 138:6 G 4:8
points, and rat strains. For example, one study
Pair-fed control 6 36:5 G 0:8 212:8 G 10:2
showed that the hippocampal BDNF content ini-
Each value is represented as mean G SEM. tially increased after 6–12 weeks of ethanol admin-
n: number of animals examined, BDNF: brain-derived neuro- istration and subsequently decreased after 24 weeks
trophic factor, GDNF: glial-derived neurotrophic factor. of ethanol administration as a liquid diet containing
8.1%–9.4% (v/v) ethanol18,19). However, in an-
other study, 6.6% (w/v) ethanol administered as a
liquid diet for 6 weeks resulted in no alterations
in the hippocampal BDNF and Trk B mRNA
of the PCR products on 2% agarose gel; a single levels20). Similarly, no change in neurotrophin pro-
band confirmed that specific amplification had been tein and mRNA levels in the rat hippocampus was
accomplished. When RT was excluded, the amplifi- observed after ethanol treatment for 28 weeks21).
cation products were not detected. The above se- The precise reasons for these discrepancies are un-
ries of procedures was performed for each sample. clear but may arise due to significant variations in
Additionally, the melting curves of the PCR prod- the ethanol treatment and the sampling times used
ucts amplified using BDNF, GDNF, and GAPDH in the various studies. One of the possible ex-
primer pairs showed a single and sharp transition. planations for this discrepancy is the differences in
This confirms that a single PCR product was pres- the rat strains. For example, the Long-Evans strain
ent, and primer dimer formation was a rare occur- of rats used by the MacLennan’ group18) may be
rence within the number of cycles required for more sensitive to ethanol in terms of hippocampal
quantification. These results provide a strong con- neurotrophin responsiveness than the Sprague-
firmation that the primers used in this study amplify Dawley rats20).
the target genes BDNF, GDNF, and GAPDH and It has been reported that GDNF protects neu-
exclude genomic DNA contamination. rons against ethanol-induced neuronal damage22).
Table 3 shows the mean G SEM ratio (10 2 ) of We hypothesized that chronic ethanol treatment
the expression levels of BDNF and GDNF mRNAs may increase the expression levels of glial cell-
to that of GAPDH mRNA in chronic ethanol-fed associated proteins, including GDNF, since glial
and pair-fed control rats. No significant differences cells are activated by various extrinsic insults. Con-
in both BDNF and GDNF mRNA levels were ob- trary to our hypothesis, the present study demon-
served between chronic ethanol-fed and pair-fed strated that chronic ethanol administration for 19
control rats at the end of ethanol treatment. weeks did not result in a significant change in the
expression level of GDNF mRNA when compared
with that in controls. Recently, it has been reported
that ethanol inhibits GDNF protein release mainly
Discussion from astrocytes but does not affect the mRNA
expression of this protin23). Our present data on
Chronic ethanol ingestion is known to cause GDNF appear to be consistent with this report.
brain damage and cognitive impairment in humans Since GDNF is mainly released from astrocytes
and laboratory animals12–17). Since various neuro- in the CNS23,24), it is likely that astrocytes are less
trophic factors are implicated in normal develop- influenced by chronic ethanol exposure. Further
ment, functional maintenance, plasticity, and re- studies are required to understand how GDNF
covery from neuronal damage, these functional might act against the neurotoxic effects of ethanol.
disorders in the hippocampus are closely related The alterations in the expression levels of BDNF
to alterations in the expression levels of various and GDNF mRNAs after ethanol administration
neurotrophins, including BDNF and GDNF. How- may be explained by several potential mechanisms.
ever, our results demonstrated that chronic ethanol One possible explanation is that GDNF and BDNF
treatment for 19 weeks resulted in no significant act in concert through common intracellular targets
alterations in the expression levels of BDNF and such as the Ras/Erk pathway and together activate
GDNF mRNAs in the rat hippocampus. the survival response to the neurotoxic effects of
Chronic Ethanol Administration and Neurotrophic Factors 5

ethanol25–29). Our data showing unaltered expres- Acknowledgements


sion levels of BDNF and GDNF mRNAs appear
to be consistent with these reports. However, it is This work was partly supported by a Grant-in-
equally likely that BDNF and GDNF act indepen- Aid for Scientific Research from the Ministry of
dently30). The precise reason for this discrepancy is Education, Culture, Sports, Science and Technol-
unclear at present. Further studies are required to ogy of Japan (B2, 16390310).
clarify this discrepancy.
Neuropathological changes in human alcoholic
patients were reviewed in detail by Harper31). The
brain weight of alcoholics is generally lower than References
that of controls, and the degree of brain atrophy
correlates with the rate and amount of alcohol 1) Ollat H, Parvez H and Parvez S. Alcohol and central neu-
consumption over the lifetime. The reduction in rotransmission. Neurochem Int 1998; 13:275–300.
2) Deitrich R, Dunwiddie T, Harris R and Erwin V. Mecha-
brain weight and volume is strongly correlated with nism of action of ethanol: initial central nervous system
a reduction in white matter volume in the pre- actions. Pharmacol Rev 1989; 41:489–537.
frontal cortex32). The mechanism for the ethanol- 3) Samson H and Harris R. Neurobiology of alcohol abuse.
induced loss of white matter volume remains un- Trends Pharmacol Sci 1992; 13:206–211.
4) Satriotomo I, Miki T, Itoh M, Ameno K, Ijiri I, Takeuchi
known at present; however, it is strongly speculated
Y. Short-term ethanol exposure alters calbindin D28k and
that both myelination and axonal integrity are in- glial fibrillary acidic protein immunoreactivity in hippo-
volved in this phenomenon to a certain extent. campus of mice. Brain Res 2000; 879:55–64.
Demyelination is one of the neuropathological 5) Maisonpierre P, Belluscio L, Friedman B, Alderson R,
changes observed in human alcoholics31). Myelin- Wiegand S, Furth M, Lindsay R and Yancopoulos G. NT-3,
associated proteins such as oligodendrocyte myelin BDNF, and NGF in the developing rat nervous system:
parallel as well as reciprocal patterns of expression. Neuron
glycoprotein (OMgp), myelin-associated glyco- 1990; 5:501–509.
protein (MAG), and Nogo are categorized as 6) Das KP, Chao SL, White LD, Haines WT, Harry GJ,
myelin-associated inhibitors and belong to the Tilson HA and Barone SJ. Differential patterns of nerve
Nogo receptor (NgR) ligand family. These proteins growth factor, brain-derived neurotrophic factor and
are expressed by oligodendrocytes and ultimately neurotrophin-3 mRNA and protein levels in developing
regions of rat brain. Neuroscience 2001; 103:739–761.
lead to degeneration following neuronal dam- 7) Lin L, Doherty D, Lile J, Bektesh S and Collins F. GDNF:
age33–37). We have recently found that ethanol ad- a glial cell line-derived neurotrophic factor for midbrain
ministration to rats for 19 weeks resulted in a sig- dopaminergic neurons. Science 1993; 260:1130–1132.
nificant decrease in the expression levels of OMgp 8) Hoffer B, Hoffman A, Bowenkamp K, Huettl P, Hudson J,
Martin D, Lin L and Gerhardt G. Glial cell line-derived
mRNA in the hippocampus (unpublished data). It
neurotrophic factor reverses toxin-induced injury to mid-
is speculated that the decreased expression level of brain dopaminergic neurons in vivo. Neurosci Lett 1994;
OMgp mRNA is attributed to the damage to the 182:107–111.
oligodendrocytes/myelin sheath induced by chronic 9) Li L, Wu W, Lin L, Lei M, Oppenheim R and Houenou L.
ethanol consumption. Our data on OMgp mRNA Rescue of adult mouse motoneurons from injury-induced
appear to be consistent with the neuropathological cell death by glial cell line-derived neurotrophic factor.
Proc Natl Acad Sci USA 1995; 92:9771–9775.
changes in alcoholics, which are characterized by 10) Lindvall O, Kokaia Z, Bengzon J, Elmér E and Kokaia
demyelination. M. Neurotrophins and brain insults. Trends Neurosci 1994;
In summary, chronic ethanol administration for 17:490–496.
19 weeks did not significantly affect the expression 11) Kuma H, Miki T, Matsumoto Y, Gu H, Li H-P, Kusaka T,
levels of BDNF and GDNF mRNAs in the rat hip- Satriotomo I, Okamoto H, Yokoyama T, Bedi K, Onishi S,
Suwaki H and Takeuchi Y. Early maternal deprivation in-
pocampus. However, the functional implications of duces alterations in brain-derived neurotrophic factor ex-
alterations in the expression levels of these mRNAs pression in the developing rat hippocampus. Neurosci Lett
are uncertain at present. It is speculated that vari- 2004; 372:68–73.
ous neurological symptoms seen in human alco- 12) Walker D and Freund G. Impairment of shuttle box
avoidance learning following prolonged alcohol consump-
holics are closely related to alterations in the ex-
tion in rats. Physiol Behav 1970; 7:773–778.
pression levels of these important neurotrophic 13) Walker D and Hunter B. Short-term memory impairment
factors. Further experiments that include parame- following chronic alcohol consumption in rats. Neuro-
ters such as different periods of ethanol adminis- psychologia 1978; 16:545–553.
tration, multiple sampling points, and various etha- 14) Charness M. Brain lesions in alcoholics. Alcohol Clin Exp
nol concentrations in the diet are required to clarify Res 1993; 17:2–11.
15) Victor M. Persistent altered mentation due to ethanol.
the effects of chronic ethanol administration on al- Neurol Clin 1993; 11:639–661.
terations in the levels of gene expression of neuro- 16) Victor M. Neurologic disorders due to alcoholism and
trophins. malnutrition. In: Joynt RJ, Griggs RC, eds, Clincial neu-
6 H. Okamoto et al.

rology: Philadelphia: Lippincott, Williams and Wilkins, 2A, and MEN 2B mutations. Biochem Biophys Res Com-
1998. mun 1994; 237:747–751.
17) Steigerwald E and Miller M. Performance by adult rats in 27) van-Weering DH and Bos JL. Glial cell line-derived neu-
sensory-mediated radial arm maze tasks is not impaired rotrophic factor induces Ret-mediated lamellipodia forma-
and may be transiently enhanced by chronic exposure to tion. J Biol Chem 1997; 272:249–254.
ethanol. Alcohol Clin Exp Res 1997; 21:1553–1559. 28) van-Weering DH and Bos JL. Signal transduction by the
18) MacLennan A, Lee N and Walker D. Chronic ethanol ad- receptor tyrosine kinase Ret. Recent Results Cancer Res
ministration decreased brain-derived neurotrophic factor 1998; 154:271–281.
gene expression in the rat hippocampus. Neurosci Lett 29) Trupp M, Scott R, Whittemore SR and Ibanez CF. Ret-
1995; 197:105–108. dependent and -independent mechanisms of glial cell line-
19) Miller M. Repeated episodic exposure to ethanol affects derived neurotrophic factor signaling in neuronal cells. J
neurotrophin content in the forebrain of the mature rat. Biol Chem 1999; 274:20885–20894.
Exp Neurol 2004; 189:173–181. 30) Feng L, Wang CY, Jiang H, Oho C, Dugich DM, Mei L and
20) Zhang L, Dhillon H, Barron S, Hicks R, Prasad R and Se- Lu B. Differential signaling of glial cell line-derived neuro-
roogy K. Effects of chronic ethanol administration on ex- trophic factor and brain-derived neurotrophic factor in
pression of BDNF and trkB mRNAs in rat hippocampus cultured ventral mesencephalic neurons. Neuroscience
after experimental brain injury. Mol Brain Res 2000; 1999; 93:265–273.
79:174–179. 31) Harper C. The neuropathology of alcohol-specific brain
21) Baek JK, Heaton MB and Walker DW. Chronic alcohol damage, or does alcohol damage the brain? J Neuropathol
ingestion: nerve growth factor gene expression and neuro- Exp Neurol 1998; 57:101–110.
trophic activity in rat hippocampus. Alcohol Clin Exp Res 32) Sullivan E and Pfefferbaum A. Diffusion tensor imaging in
1994; 18:1368–1376. normal aging and neuropsychiatric disorders. Eur J Radiol
22) McAlhany RE Jr, West JR and Miranda RC. Glial- 2003; 45:244–255.
derived neurotrophic factor rescues calbindin-D28k- 33) Chen M, Huber A, van der Haar M, Frank M, Schnell L,
immunoreactive neurons in alcohol-treated cerebellar ex- Spillmann A, Christ F and Schwab M. Nogo-A is a myelin-
plant cultures. J Neurobiol 1997; 20:835–847. associated neurite outgrowth inhibitor and an antigen for
23) McAlhany RE Jr, Miranda RC, Finnell RH and West JR. monoclonal antibody IN-1. Nature 2000; 403:434–439.
Ethanol decreases Glial-Derived Neurotrophic Factor 34) GrandPre T, Nakamura F, Vartanian T and Strittmatter
(GDNF) protein release but not mRNA expression and SM. Identification of the Nogo inhibitor of axon regenera-
increases GDNF-stimulated Shc phosphorylation in the tion as a Reticulon protein. Nature 2000; 403:439–444.
developing cerebellum. Alcohol Clin Exp Res 1999; 35) Prinjha R, Moore S, Vinson M, Blake S, Morrow R, Chris-
23:1691–1697. tie G, Michalovich D, Simmons D and Walsh F. Inhibitor of
24) Tomac A, Lindqvist E, Lin L, Ogren S, Young D, Hoffer B neurite outgrowth in humans. Nature 2000; 403:383–384.
and Olson L. Protection and repair of the nigrostriatal 36) Domeniconi M, Cao Z, Spencer T, Sivasankaran R, Wang
dopaminergic system by GDNF in vivo. Nature 1995; K, Nikulina E, Kimura N, Cai H, Deng K, Gao Y, He Z
373:335–339. and Filbin M. Myelin-associated glycoprotein interacts with
25) Kaplan DR and Stephens RM. Neurotrophin signal trans- the Nogo 66 receptor to inhibit neurite outgrowth. Neuron
duction by the Trk receptor. J Neurobiol 1994; 25:1404– 2002; 35:283–290.
1417. 37) Wang KC, Koprivica V, Kim JA, Sivasankaran R, Guo Y
26) Ohiwa M, Murakami H, Iwashita T, Asai N, Iwata Y, Imai and Neve RL, Z. H. Oligodendrocyte-myelin glycoprotein
T, Funahashi H, Takagi H and Takahashi M. Character- is a Nogo receptor ligand that inhibits neurite outgrowth.
ization of Ret-Shc-Grb2 complex induced by GDNF, MEN Nature 2002; 417:941–944.

You might also like