You are on page 1of 6

ABSTRACT: Glial cell line–derived neurotrophic factor (GDNF) is pro-

duced by skeletal muscle and affects peripheral motor neurons. Elevated


expression of GDNF in skeletal muscle leads to hyperinnervation of neuro-
muscular junctions, whereas postnatal administration of GDNF causes syn-
aptic remodeling at the neuromuscular junction. Studies have demonstrated
that altered physical activity causes changes in the neuromuscular junction.
However, the role played by GDNF in this process in not known. The ob-
jective of this study was to determine whether changes in neuromuscular
activity cause altered GDNF content in rat skeletal muscle. Following 4
weeks of walk-training on a treadmill, or 2 weeks of hindlimb unloading,
soleus, gastrocnemius, and pectoralis major were removed and analyzed for
GDNF content by enzyme-linked immunosorbant assay. Results indicated
that walk-training is associated with increased GDNF content. Skeletal
muscle from hindlimb-unloaded animals showed a decrease in GDNF in
soleus and gastrocnemius, and an increase in pectoralis major. The altered
production of GDNF may be responsible for activity-dependent remodeling
of the neuromuscular junction and may aid in recovery from injury and dis-
ease.
© 2002 Wiley Periodicals, Inc. Muscle Nerve 26: 206–211, 2002

GDNF IS REGULATED IN AN ACTIVITY-DEPENDENT


MANNER IN RAT SKELETAL MUSCLE
ERICA A. WEHRWEIN, MS, ERIC M. ROSKELLEY, BS, and JOHN M. SPITSBERGEN, PhD

Department of Biological Sciences, Western Michigan University, Kalamazoo, Michigan 49008, USA

Accepted 27 March 2002

Most neurons require a constant supply of neuro- Although several recent studies have examined
trophic factors for growth, maintenance of cell phe- the regulation of GDNF expression in neurons and
notype, and possibly continued survival in adult glial cells,2,16,29 very little is known about processes
organisms. Alterations in neurotrophic factor pro- involved in the regulation of GDNF expression in
duction, release, or biological effects may have pro- muscle. Studies examining neurotrophic factor ex-
nounced effects on nervous system structure and pression in smooth muscle have shown that a variety
function. Although a variety of neurotrophic factors of stimuli can influence its expression, including ex-
have been identified that exert effects on motor neu- posure to neurotransmitters 22 and mechanical
rons,16 the findings that glial cell line–derived neu- stretch.19 If alterations in mechanical activity affect
rotrophic factor (GDNF) is present in mature skel- skeletal muscle GDNF expression, then changes in
etal muscle23 and alters structure and function in physical activity could affect motor neuron structure
mature motor neurons,31 makes GDNF an excellent and function indirectly through changes in GDNF
candidate as a neurotrophic molecule controlling expression.
motor neuron plasticity in adult organisms. Increased physical activity increases size and de-
gree of branching of motor nerve terminals at neu-
romuscular junctions of type I fibers,1,10 increases
expression of acetylcholinesterase,24 increases ex-
pression of acetylcholine receptor,9 and increases
Abbreviations: BSA, bovine serum albumin; EDTA, ethylenediamine-
tetra-acetic acid; ELISA, enzyme-linked immunosorbant assay; GDNF, size and density of pre- and post-synaptic membrane
glial cell line–derived neurotrophic factor; PBS, phosphate-buffered saline specializations.25,27 There are differential effects on
Key words: exercise; GDNF; neuromuscular junction; neurotrophic fac-
tor; plasticity soleus neuromuscular junction morphology based
Correspondence to: J. Spitsbergen; e-mail: john.spitsbergen@wmich. on intensity of training.10 Animals trained at either
edu
high or low intensity demonstrated neuromuscular
© 2002 Wiley Periodicals, Inc.
Published online 10 June 2002 in Wiley InterScience (www.
junction hypertrophy; however, high-intensity
interscience.wiley.com). DOI 10.1002/mus.10179 trained animals had more dispersed synapses,

206 Activity Alters GDNF in Muscle MUSCLE & NERVE August 2002
greater total length of branching, higher average All animals were monitored daily and body
length per branch, and greater number of secondary weights were measured to ensure positive weight
branches than the low-intensity group.10 gain. Throughout the 4-week experimental period,
Hindlimb unloading and space flight lead to control animals remained in their cages and partici-
muscle atrophy,28 neuromuscular junction atrophy,3 pated in normal ambulation. Walk-trained animals
and neuromuscular weakness.11 The atrophic re- were treadmill-trained using the following param-
sponse of skeletal muscle in the soleus and gastroc- eters: 15 min/day at 0% incline (three 5-min inter-
nemius is maximal after 2 weeks of suspension or vals with 8-min rest period between trials) at 21.3
space flight.20 Post-flight, there appears to be a de- m/min, 5 days/week for 4 weeks. This protocol was
creased number of synaptic vesicles in the nerve ter- adapted from Tomas et al.26 This low-intensity exer-
minal and mitochondrial swelling, which has been cise, with breaks between exercise periods, was de-
interpreted as the first stage in axonal degenera- signed to utilize primarily low-threshold motor neu-
tion.4 It has been demonstrated that muscle activity, rons innervating type I skeletal muscle fibers.
both frequency and amplitude, are decreased during Hindlimb-unloaded animals were subjected to hind-
hindlimb unloading.5 limb unloading for 2 weeks and then sacrificed as
We tested the hypothesis that GDNF expression described below.
in skeletal muscle is regulated by the level of neuro- The 2-week time course was selected based on
muscular activity. We predicted that increased physi- data indicating that 14 days of unweighting resulted
cal activity would lead to an increase in GDNF ex- in maximal atrophy of soleus and gastrocnemius and
pression, and decreased physical activity to a that neuromuscular junction modifications were also
decrease in GDNF expression. If GDNF expression is noted at this time point.20 During this time, animals
controlled by the level of neuromuscular activity, remained in their cages and were not allowed to rest
then the alterations in motor neuron structure and their hindlimbs on the floor of the cage.
function seen with changes in physical activity may Each training day, animals in the exercise group
be driven by changes in GDNF expression. were allowed to warm-up for 1 min at 5.3 m/min (0.2
mph) on the treadmill prior to beginning the exer-
MATERIALS AND METHODS cise. Training was performed using a simple escape
contingency. Water served as the punishment in the
Subjects. All animal experiments were performed escape contingency when subjects failed to perform
in accordance with the “Guide for the Care and Us- at a satisfactory level. When a subject stopped walk-
age of Laboratory Animals” (National Research ing, it was sprayed with water. Animals were sacri-
Council) and experiments were approved by our In- ficed by CO2-euthanization prior to thoracotomy 24
stitutional Animal Care and Usage Committee. h after the final bout of exercise. Age-matched con-
Twenty-one healthy, adult (8 weeks) male Sprague- trol animals were sacrificed on the same day.
Dawley rats (Charles River, Portage, Michigan) were
randomly assigned to sedentary control (n = 7), walk- Tissue Collection and Processing. Selected skeletal
trained (n = 7), or hindlimb-unloaded (n = 7) muscles (soleus, gastrocnemius, and pectoralis ma-
groups. Weights at the onset of the experiment jor) were removed and flash frozen upon contact
ranged from 230.1 g to 250.7 g with an average of with dry ice. Frozen tissue was cut into serial cross-
240.3 ± 1.7 g. Control and walk-trained animals were sections weighing 0.5 g or less and stored at −80°C.
housed one or two per cage in Nalgene cages with Muscles were removed bilaterally. The left-sided
access to food and water ad libitum. Animals were muscles were used for protein assay and the right-
housed with another animal in the same treatment sided muscles for immunocytochemistry. Just prior
group unless there were no treatment-matched ani- to processing, frozen muscle samples were dipped
mals available, in which case the animal was housed into liquid nitrogen and then pulverized on a metal
alone. Hindlimb-unloaded rats were housed singly in block chilled on dry ice. The pulverized muscle was
modified rabbit cages. Hindlimb suspension was ac- then suspended in 0.1 M phosphate-buffered saline
complished by taping the tail of the rat to the ceiling (PBS: 0.225 M NaCl, 0.02 M NaH2PO4, 0.08 M
of a rabbit cage, such that the hindlimbs of the rat Na2HPO4) containing 0.1% Tween-20, 0.05% bovine
were elevated off the bottom of the cage.18 Animals serum albumin (BSA), aprotinin (Sigma, St. Louis,
were suspended so that access to food and water was Missouri), 0.2 mM benzamidine, 0.01 mM benzetho-
assured. Animals were kept on a 12:12 h light-dark nium chloride, and 0.2 mM ethylenediaminetetra-
cycle in a room with regulated temperature (22– acetic acid (EDTA).7 The suspension was chilled on
24°C). wet ice while being homogenized for 30 s using a

Activity Alters GDNF in Muscle MUSCLE & NERVE August 2002 207
variable speed Tissue Tearor (Biospec Products, 1-h incubation in primary antibody at room tempera-
Inc., Bartlesville, Oklahoma). Homogenate was cen- ture. Slides were then treated with a streptavidin-
trifuged at 13,000g and the supernatant was analyzed linked probe (Alexa 488, Molecular Probes, Eugene,
using an enzyme-linked immunosorbant assay Oregon) diluted in PBS, at 37°C, for 30 min. Images
(ELISA) specific for GDNF. were captured using confocal microscopy (Zeiss Co.,
Thornwood, New York) at an excitation wavelength
Enzyme-Linked Immunosorbant Assay. Assay plates of 495 nm and emission wavelength of 519 nm. Sec-
(96-well NUNC-Immuno, Nalgene Nunc Interna- tions were viewed in 2 µm optical slices at 20× mag-
tional, Rochester, New York) were incubated with a nification. Negative control images were obtained by
monoclonal antibody raised against GDNF (R&D omitting incubation with primary antibody and uti-
Systems, Minneapolis, Minnesota) overnight in a hu- lizing a streptavidin-linked fluorescent probe.
midified chamber. Remaining sites were blocked
with BSA (1.0%) for 1 h at room temperature. Plates GDNF and Di-I Colocalization. GDNF-stained skel-
were rinsed three times and muscle sample homog- etal muscle sections were prepared as described
enate or GDNF standard (R&D Systems) was added above and then stained with 1,1⬘-dioctadecyl-
to each well. All samples and standards were run in 3,3,3⬘,3⬘-tetramethylindocarbocyanine perchlorate/
quadruplicate. GDNF standard (2 µg/ml) was di- DiIC18 (Di-I) (Molecular Probes). A stock Di-I solu-
luted in sample buffer to prepare a standard curve tion of 10 mg/ml was prepared in 50:50
between 1,000 pg/ml and 2 pg/ml. Internal control ethanol:dimethyl sulfoxide. A working concentra-
wells were used that contained only sample buffer. tion of 1:100 was prepared from the stock and di-
Sample incubation was done in a humidified cham- luted in PBS. Dilute Di-I (100 µl) was placed directly
ber for 2 h at room temperature. Following incuba- on the slide and cover slipped. The slide was allowed
tion of the sample or standard, the wells were washed
to incubate at room temperature for 15 min. Follow-
and incubated with an anti-GNDF antibody conju-
ing incubation, the sections were rinsed three times
gated to biotin (R&D Systems) for 2 h at room tem-
for 5 min in PBS. Sections labeled with anti-GDNF
perature. The wells were then washed three times
and Di-I were visualized using confocal microscopy
and horseradish peroxidase conjugated to streptavi-
as described above, at an excitation wavelength of
din was added for 20 min at room temperature. The
535 nm and an emission wavelength of 590 nm for
wells were washed three times, and 3,3⬘,5,5⬘-
Di-I visualization.
tetramethylbenzidine substrate was added and al-
lowed to incubate approximately 30 min at room
Statistical Analysis. Comparisons between control
temperature. The reaction was stopped with 0.1 M
phosphoric acid, and plates were read at 450 nm and treatment were made using a Student’s t-test.
using a microplate spectrophotometer. Standard For all tests, significance was set to P ⱕ 0.05. All data
curves were derived from known GDNF samples. values are reported as the mean ± standard error of
Muscle extract data was reported as picograms of the mean.
GDNF per gram of tissue wet weight.
RESULTS

Immunocytochemistry. Skeletal muscle samples Gross Observations. Animal body weights were not
were removed from the right side of the animal and significantly different at the onset of the experiment;
immediately frozen in 2-methyl-butane chilled on however, weights of walk-trained (407.0 ± 11.3) and
dry ice. Tissue was stored at −80°C until sectioned. hindlimb-unloaded animals (414.86 ± 6.9) were sig-
Whole muscle was divided into three sections. Sec- nificantly lower (P ⱕ 0.05) than control animals
tions for imaging were taken from the middle third (439.98 ± 8.9) at the end of the 4-week training pe-
of the whole muscle. Cryostat sections (50 µm; cross riod or 2-week unloading period. All animals showed
section) were mounted on slides and then affixed by positive weight gain throughout the experiment.
placing slides in a vacuum-sealed container for 4 h. Measurements of individual muscle weights were
Sections were subsequently fixed using 4% para- also taken. The weights of soleus (0.70 ± 0.6 g), gas-
formaldehyde diluted in PBS at room temperature trocnemius (0.77 ± 0.16 g) and pectoralis major
for 15 min. Samples were blocked using 1% BSA in (0.79 ± 0.1 g) from control rats were not different
PBS for 20 min then rinsed for 10 min in wash from walk-trained rats (0.75 ± 0.06 g, 0.73 ± 0.16 g,
buffer. Biotinylated primary polyclonal antibody and 0.71 ± 0.1 g, respectively) or from hindlimb-
raised against GDNF (R&D Systems) was used in a unloaded rats (0.63 ± 0.06 g, 0.61 ± 0.16 g, and 0.82
1:500 dilution in PBS. A 15-min PBS wash followed a ± 0.1 g, respectively).

208 Activity Alters GDNF in Muscle MUSCLE & NERVE August 2002
GDNF Content in Control Muscles. Immunocyto- (Fig. 2). Hindlimb unloading resulted in a signifi-
chemical staining was performed on gastrocnemius cant decrease in GDNF content in soleus and gas-
from control animals to ensure that there was a mea- trocnemius compared to controls (P < 0.05) (Fig. 2).
surable amount of GDNF in control skeletal muscle. By contrast, hindlimb unloading resulted in a signifi-
Immunoreactivity for GDNF was observed in the rat cant increase in GDNF content in pectoralis major
gastrocnemius muscle from control animals. GDNF (P < 0.05) compared to control (Fig. 2).
staining in cross sections appeared to be localized to
the membrane (Fig. 1). Membrane localization was DISCUSSION
confirmed by demonstrating colocalization of GDNF
and Di-I to the membrane of rat skeletal muscle. The Our experiments showed that GDNF content in skel-
pattern of membrane-associated GDNF is consistent etal muscle was increased following 4-weeks of walk-
with that previously described.23 Control sections training. Skeletal muscle from hindlimb-suspended
were stained with streptavidin-linked fluorescent animals showed a significant decrease in GDNF in
probe in the absence of primary antibody. Images soleus and gastrocnemius, and an increase in GDNF
obtained in the absence of primary antibody showed in pectoralis major. These findings suggest that
low levels of background staining. GDNF is modulated by activity level in vivo. It re-
mains to be seen whether changes in GDNF expres-
Activity-Dependent Alterations in GDNF. In order sion with activity play a role in the changes in neu-
to study activity-dependent changes in trophic fac- romuscular junction architecture observed following
tor, we selected two hindlimb locomotor muscles, increased motor activity.
soleus (type I) and gastrocnemius (mixed fiber GDNF is a survival factor for peripheral nerves
type). Soleus is a primary load-bearing muscle and and muscle.12 It is important in motor and sensory
was used to examine the effects of hindlimb unload- neuron development,30 but its role in adult rats is
ing as well. Pectoralis major, a mixed fiber type unclear. Results of a previous study6 showed that
muscle, was selected as it continues to bear weight in skeletal muscle from rat contains low levels of GDNF
the hindlimb unloading studies. When GDNF con- mRNA, which led others to suggest a limited role for
tent of soleus, gastrocnemius, and pectoralis major GDNF in adult skeletal muscle. We have demon-
from walk-trained animals was measured using an strated immunocytochemically and utilizing ELISA
ELISA, we found an activity-dependent, significant that GDNF protein is indeed present in measurable
increase in GDNF (P < 0.05) compared to controls amounts in adult rat skeletal muscle. Using whole

FIGURE 1. Localization of GDNF protein in rat skeletal muscle from control animals. Left panel shows staining in the presence of
anti-GDNF primary antibody that appears to be localized to the membrane. (bar = 50 µm). Right panel shows colocalization of GDNF and
Di-I staining to the membrane of skeletal muscle cells (bar = 50 µm).

Activity Alters GDNF in Muscle MUSCLE & NERVE August 2002 209
tylcholinesterase in cultured myotubes.13 Nifedipine
treatment partially blocks the effects of stretch on
acetylcholinesterase expression, indicating that
stretch induces calcium influx through voltage-gated
calcium channels.13 Thus, increased mechanical ac-
tivity may be responsible for some of the increased
GDNF levels observed in all muscles from walk-
trained rats and pectoralis major from hindlimb-
unloaded rats, whereas decreased mechanical activ-
ity may mediate the decrease in GDNF content of
hindlimb muscle from hindlimb-unloaded rats.
Data indicating that neurotransmitters modulate
neurotrophic factor expression comes from studies
in vascular and bladder smooth muscle cells in cul-
ture. In these studies, neurotransmitters from sym-
pathetic neurons alter production of nerve growth
factor by smooth muscle cells in culture.8,21 Indirect
evidence that motor neurotransmitters exert a nega-
tive influence on trophic factor production by skel-
etal muscle comes from studies performed on devel-
oping chick neuromuscular junctions. Oppenheim
et al.17 demonstrated that blockade of nicotinic ace-
tylcholine receptors on skeletal muscle enhanced
the ability of skeletal muscle to provide trophic sup-
FIGURE 2. Effect of hindlimb unloading (A) and walk-training (B)
on skeletal muscle GDNF content. (A) GDNF content is signifi- port to innervating motor neurons. Direct evidence
cantly increased in skeletal muscle following 4 weeks of walk- that neurotransmitters exert a negative influence on
training as compared to sedentary control. White bars represent GDNF expression in skeletal muscle has been ob-
control and black bars represent walk-training. (B) Soleus and tained from studies demonstrating that denervated
gastrocnemius showed a significant decrease in GDNF content
as compared to matched muscle from sedentary controls,
skeletal muscle contains elevated levels of GDNF
whereas pectoralis major showed a significant increase in GDNF mRNA.14 Cell culture data from this laboratory22 in-
as compared to sedentary control. White bars represent control dicate that acetylcholine inhibits GDNF secretion
and black bars represent hindlimb unloading. Values are means from L6 skeletal muscle cells.
± SEM of skeletal muscle from seven animals. The asterisk in-
Hindlimb unloading leads to a decrease in neu-
dicates a value significantly different than control (P ⱕ 0.05).
romuscular junction complexity3,4,11,20 and we hy-
pothesized that this may be due to a decrease in
muscle homogenate, GDNF released from skeletal trophic factor expression. Studies have shown that
muscle cannot be distinguished from GDNF released overexpression of GDNF leads to hyperinnervation
from adherent Schwann cells. However, the findings of neuromuscular junctions.15 It is possible that a
from the L6 skeletal muscle cell line in culture22 and decrease in GDNF expression leads to a decrease in
of immunocytochemical studies 23 indicate that innervation. In our study, soleus and gastrocnemius
GDNF is produced by skeletal muscle cells. showed a decrease in GDNF production following
This study has shown differential effects on tro- hindlimb unloading. The increase in GDNF seen in
phic factor production based on activity level. Fac- the pectoralis major may be due to an increased load
tors that may mediate trophic factor expression in- on the muscle from the altered body position
clude mechanical activity and neurotransmitter achieved with hindlimb unloading.
release. Mechanical activity, in the form of stretch, Activity-dependent regulation of trophic factors
appears to be a positive stimulus for trophic factor has far-reaching implications. If trophic factors are
production in vascular and bladder smooth muscle regulated in an activity-dependent manner, then
cells. Cyclic and static stretch increase production conditions such as sedentary lifestyle, immobiliza-
and release of nerve growth factor in both bladder tion due to injury or illness, age-related decreases in
and vascular smooth muscle cells of rats8 by activa- activity, hindlimb suspension, neuromuscular injury
tion of protein kinase C signaling pathways.19 Me- or illness, and exposure to microgravity may lead to
chanical stimulation increases the expression of ace- dramatic changes in trophic factor expression and

210 Activity Alters GDNF in Muscle MUSCLE & NERVE August 2002
subsequent neuromuscular junction remodeling. 13. Hubatsch DA, Jasmin BJ. Mechanical stimulation increases
expression of acetylcholinesterase in cultured myotubes. Am
Trophic factor expression may play a prominent role J Physiol 1997;273:2002–2009.
in mediating these changes. 14. Lie DC, Weis J. GDNF expression is increased in denervated
human skeletal muscle. Neurosci Lett 1998;250:87–90.
This work was supported by NIH grant 1 R15 HL60240-01, The
15. Nguyen QT, Parsadanian AS, Snider WD, Lichtman JW. Hy-
Faculty Research and Support Fund of Western Michigan Univer- perinnervation of neuromuscular junctions caused by GDNF
sity, and a Grant-In-Aid from the American Heart Association overexpression in muscle. Science 1998;279:1725–1729.
(Michigan Affiliate). Preliminary results were presented at the 16. Oppenheim R. Neurotrophic survival molecules for motoneu-
Integrative Biology of Exercise Conference, Portland, Maine, Sep- rons: an embarrassment of riches. Neuron 1996;17:195–197.
tember 2000. The authors acknowledge the advice and assistance 17. Oppenheim RW, Prevette D, D’Costa A, Wang S, Houenou
of Drs. Christine Byrd, John Jellies, and William Jackson, Western LJ, McIntosh JM. Reduction of neuromuscular activity is re-
Michigan University. We greatly appreciate the technical assis- quired for the rescue of motoneurons from naturally occur-
tance of Berta C.R. Cohen. We also thank Matthew P. DeVries, ring cell death by nicotinic-blocking agents. J Neurosci 2000;
Angela Lim, Marisa Hart, and Judy Martin for their assistance in 20:6117–6124.
tissue processing, and Drs. Rob Eversole and John Stout in the 18. Park E, Schultz E. A simple hindlimb suspension apparatus.
Biological Imaging Center for assistance with confocal imaging. Aviat Space Environ Med 1993;64:401–404.
19. Persson K, Sando JJ, Tuttle JB, Steers WD. Protein kinase C in
cyclic stretch-induced nerve growth factor production by uri-
nary tract smooth muscle cells. Am J Physiol 1995;269:
REFERENCES
C1018–C1024.
1. Andonian MH, Fahim MA. Effects of endurance exercise on 20. Roy RR, Baldwin KM, Edgerton VR. Response of the neuro-
the morphology of mouse neuromuscular junctions during muscular unit to spaceflight: what has been learned from the
ageing. J Neurocytol 1987;16:589–599. rat model. Exerc Sport Sci Rev 1996;24:399–425.
2. Appel E, Kolman O, Kazimirsky G, Blumberg PM, Brodie C. 21. Spitsbergen JM, Stewart JS, Tuttle JB. Altered regulation of
Regulation of GDNF expression in cultured astrocytes by in- nerve growth factor secretion by cultured VSMCs from hyper-
flammatory stimuli. Neuroreport 1997;8:3309–3312. tensive rats. Am J Physiol 1995;269:H621–H628.
3. Baranski S, Marciniak M. Morphometric ultrastructural analy- 22. Spitsbergen JM, Sloma MM, Chadalavada SC. Regulation of
sis of muscle fibers and neuromuscular junction in rats kept expression of glial cell line derived neurotrophic factor
under conditions of weightlessness for 21 days. Mater Med Pol (GDNF) in cultured L6 skeletal muscle cells. Soc Neurosci
1990;22:258–262. Abstr 1999;25:1273.
4. Baranski S, Baranska W, Marciniak M, Ilyina-Kakueva EI. Ul- 23. Suzuki H, Hase A, Miyata Y, Arahata K, Akazawa C. Prominent
tasonic investigations of the soleus muscle after space flight expression of glial cell line derived neurotrophic factor in
on the Biosputnik 936. Aviat Space Environ Med 1979;50: human skeletal muscle. J Comp Neurol 1998;402:303–312.
930–934. 24. Sveistrup H, Chan RY, Jasmin BJ. Chronic enhancement of
5. Bonen A, Blewett C, McDermott JC, Elder GC. A model for neuromuscular activity increases acetylcholinesterase gene ex-
nonexercising hindlimb muscles in exercising animals. Can pression in skeletal muscle. Am J Physiol 1995;269:C856–
J Physiol Pharmacol 1990;68:914–921. C862.
6. Choi-Lundberg DL, Bohn MC. Ontogeny and distribution of 25. Tomas J, Fenoll MR, Santafe M, Batlle J, Mayayo E. Motor
glial cell line-derived neurotrophic factor (GDNF) mRNA in nerve terminal morphologic plasticity induced by small
rat. Brain Res Dev 1995;85:80–88. changes in the locomotor activity of the adult rat. Neurosci
7. Clemow DB, Spitsbergen JM, Stewart JS, McCarty R, Steers Lett 1989;106:137–140.
WD, Tuttle JB. Altered NGF regulation may link a genetic
26. Tomas J, Batlle J, Fenoll MR, Santafe M, Lanuza MA. Activity-
predisposition for hypertension with hyperactive voiding.
dependent plastic changes in the motor nerve terminals of
J Urol 1999;161:1372–1377.
the adult rat. Biol Cell 1993;79:133–137.
8. Clemow DB, Steers WD, Tuttle JB. Stretch-activated signaling
of nerve growth factor secretion in bladder and vascular 27. Tomas J, Santafe M, Lanuza MA, Fenoll-Brunet MR. Physi-
smooth muscle cells from hypertensive and hyperactive rats. ological activity-dependent ultrastructural plasticity in normal
J Cell Physiol 2000;183:289–300. adult rat neuromuscular junctions. Biol Cell 1997;89:19–28.
9. Desaulniers P, Lavoie PA, Gardiner PF. Endurance training 28. Vandenburgh H, Chromiak J, Shansky J, Del Tatto M, Le-
increases acetylcholine receptor quantity at neuromuscular maire J. Space travel directly induces skeletal muscle atrophy.
junctions of adult rat skeletal muscle. Neuroreport 1998;9: FASEB J 1999;13:1031–1038.
3549–3552. 29. Verity AN, Wyatt TL, Lee W, Hajos B, Baecker PA, Eglen RM,
10. Deschenes MR, Maresh CM, Crivello JF, Armstrong LE, Krae- Johnson RM. Differential regulation of glial cell line-derived
mer WJ, Covault J. The effects of exercise training of different neurotrophic factor (GDNF) expression in human neuroblas-
intensities on neuromuscular junction morphology. J Neuro- toma and glioblastoma cell lines. J Neurosci Res 1999;55:
cytol 1993;22:603–615. 187–197.
11. Fujii MD, Patten BM. Neurology of microgravity and space 30. Wright DE, Snider D. Focal expression of glial cell line-
travel. Neurol Clin 1992;10:999–1013. derived neurotrophic factor in developing mouse limb bud.
12. Henderson CE, Phillips HS, Pollock RA, Davies AM, Lemeulle Cell Tissue Res 1996;286:209–217.
C, Armanini M, Simmons L, Moffet B, Vandlen RA, Simpson 31. Yan Q, Matheson C, Lopez OT. In vivo neurotrophic factor
LC. GDNF: a potent survival factor for motoneurons present effects of GDNF on neonatal and adult facial motor neurons.
in peripheral nerve and muscle. Science 1994;266:1062–1064. Nature 1995;373:341–344.

Activity Alters GDNF in Muscle MUSCLE & NERVE August 2002 211

You might also like