Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Journal of Neuropathology and Experimental Neurology Vol. 63, No.

11
Copyright q 2004 by the American Association of Neuropathologists November, 2004
pp. 1144 1154

Ganglion-Specific Patterns of Diabetes-Modulated Gene Expression Are Established in


Prevertebral and Paravertebral Sympathetic Ganglia Prior to the Development of
Neuroaxonal Dystrophy

STEVEN L. CARROLL, MD, PHD, STEPHANIE J. BYER, BS, DENISE A. DORSEY, BA, MARK A. WATSON, MD, PHD,
AND ROBERT E. SCHMIDT, MD, PHD

Abstract. In both humans and animal models, diabetic sympathetic autonomic neuropathy is associated with the selective
development of markedly enlarged distal axons and nerve terminals (neuroaxonal dystrophy, NAD). NAD occurs in the
prevertebral superior mesenteric and celiac ganglia (SMG-CG), but not in the paravertebral superior cervical ganglion (SCG).
To identify molecular differences between these ganglia that may explain their selective vulnerability to NAD, we have
examined global gene expression patterns in control and diabetic rat sympathetic ganglia before and after the onset of structural
evidence of NAD. As predicted, major differences in transcriptional profiles exist between SCG and SMG-CG in normal
young adult animals including, but not limited to, known differences in neurotransmitter-related gene expression. Gene ex-
pression patterns of diabetic SMG-CG and SCG, prior to the development of NAD lesions, also differ from their age-matched
non-diabetic counterparts. However, diabetes has ganglion-specific effects on gene expression; of approximately 110 transcripts

Downloaded from http://jnen.oxfordjournals.org/ by guest on March 21, 2016


that were differentially expressed between diabetic and control sympathetic ganglia, only 5 were differentially expressed as
a result of diabetes in both SCG and SMG-CG. Genes involving synapse and mitochondrial structure and function, oxidative
stress, and glycolysis were highly represented in the differentially expressed gene set. Differences in the number of synapse-
related gene alterations in diabetic SMG-CG (18 genes) versus SCG (2 genes) prior to the onset of NAD may also well
explain the selective development of NAD in the SMG-CG. These results provide support for the specificity of diabetes-
modulated gene expression for selected neuronal subpopulations of sympathetic noradrenergic neurons.

Key Words: Diabetes; Gene microarray; Neuroaxonal dystrophy; Sympathetic autonomic neuropathy.

INTRODUCTION STZ-induced diabetes demonstrates that NAD develops


Diabetic autonomic neuropathy, a condition affecting most reproducibly in the distal segments of long norad-
the sympathetic and parasympathetic nervous systems, renergic axons innervating the intramural submucosal
results in significant patient morbidity and substantially and myenteric ganglia of the ileum. Dystrophic changes
increased mortality (1–3). The neuropathologic hallmark do not develop in the equally lengthy nerves innervating
of diabetic sympathetic autonomic neuropathy is neu- arteries and veins within nearby neurovascular arcades
roaxonal dystrophy (NAD), a distinctive and unambigu- (4). The distinctive morphologic changes of NAD first
ous distal axonopathy resulting in markedly enlarged dis- become evident in rat superior mesenteric and celiac gan-
tal axons and nerve terminals (4). Intraganglionic glia (SMG-CG) approximately 3 months after the onset
dystrophic axons are thought to represent aberrant intra- of diabetes and fail to develop in the superior cervical
ganglionic sprouts that occur in the absence of significant ganglia (SCG), even in animals with diabetes of long
neuron loss and are hypothesized to produce dysfunction duration (6). It has been suggested that the differential
and loss of ganglionic synapses. Rats with streptozotocin development of NAD in nerves projecting to the ileum
(STZ)-induced diabetes mellitus, like human diabetic pa- and those innervating neurovascular arcades reflects their
tients, also develop pathologically distinctive degenerat- origin from prevertebral and paravertebral sympathetic
ing, regenerating, and markedly swollen dystrophic axons ganglia, respectively (4, 7, 8). Consistent with this hy-
and nerve terminals that are not accompanied by neuron pothesis, sympathetic ganglia are not equally affected by
or axon loss (5, 6). NAD in human diabetes (9), with diabetic SMG-CG
Examination of the postganglionic sympathetic axons showing marked NAD and comparable changes being
projecting to a variety of endorgans in rats with chronic virtually absent in the SCG. Prevertebral and paraverte-
bral sympathetic ganglia also differ in their response to
From Department of Pathology (SLC, SJB), The University of Ala- aging (9, 10), neonatal administration of NGF antisera or
bama School of Medicine, Birmingham, Alabama; and Department of
6-hydroxydopamine (7, 11), clonidine withdrawal (12),
Pathology and Immunology (DAD, MAW, RES), Washington Univer-
sity School of Medicine, St. Louis, Missouri. speed of axonal regeneration after experimental injury
Correspondence to: Robert E. Schmidt, MD, PhD, Washington Uni- (8), and guanethidine sympathectomy (11). These obser-
versity School of Medicine Division of Neuropathology, Department of vations indicate that there are major differences between
Pathology and Immunology, 660 South Euclid Avenue, Box 8118, St.
prevertebral and paravertebral sympathetic ganglia, some
Louis, MO 63110-1093. E-mail: reschmidt@pathology.wustl.edu
Support was provided by NIH awards R01 AG10299, R37 DK19645, of which may predispose the prevertebral ganglia to the
and R01 NS37514. development of pathologies such as NAD.

1144
GENE EXPRESSION IN DIABETIC GANGLIA 1145

Some of the distinctions between prevertebral and Health and the Washington University Committee for the Hu-
paravertebral sympathetic ganglia have been defined. mane Care of Laboratory Animals. Rats were killed 2, 6, or 17
Studies of the peripheral projections, neurotrophin sen- weeks after the onset of diabetes. At each time point, right and
left SCG, SMG, and CG from 11 to 12 diabetic rats were re-
sitivity, and function of rat SCG demonstrate that this
moved, cleaned of adherent tissue, and frozen on dry ice. For
ganglion receives presynaptic inputs from the interme-
each time point, SCG from all animals were pooled together,
diolateral column of the spinal cord and then transmits as were SMG and CG. Ganglia from 11 to 12 control rats were
signals to multiple postsynaptic targets. By comparison, similarly dissected, pooled, and frozen. The microarray data
prevertebral sympathetic ganglia have a more complex reported in this study reflect the results of 3 separate experi-
presynaptic input and a greater degree of neuronal het- ments (2 experiments performed at 2 weeks after the onset of
erogeneity (13, 14). Analyses of the effects of surgical diabetes and 1 performed 6 weeks after the onset of diabetes),
denervation and deafferentation and the expression of each utilizing a pool of ganglia from 11 to 12 animals. Initial,
neurotransmitters, their synthetic enzymes, and a variety independent analyses of microarray data from ganglia collected
of neuropeptides indicate that prevertebral sympathetic 2 and 6 weeks after the induction of diabetes showed that the
transcriptional profile of each ganglion type was essentially
ganglia are innervated by fibers projecting not only from
identical at both of these time points. Therefore, for the pur-
the intermediolateral column of the spinal cord, but also poses of this study analysis, we elected to treat all 3 experi-
from dorsal root ganglia, other sympathetic ganglia, the ments as identical ‘‘prelesion diabetic’’ time points.
vagus nerve, and retrogradely from the intramural my-

Downloaded from http://jnen.oxfordjournals.org/ by guest on March 21, 2016


enteric ganglia of the alimentary tract. Prevertebral sym- High Density Oligonucleotide Microarray Hybridization
pathetic ganglia also contain nerve terminals representing and Scanning
intraganglionic projections from neighboring neurons, a Total RNA was isolated from ganglia using TRIzol reagent
source that may become particularly relevant in patho- (Invitrogen, Carlsbad, CA) as recommended by the manufac-
logic situations. Neurons within prevertebral sympathetic turer. Double-stranded cDNA was synthesized from 20 mg of
ganglia likewise differ from paravertebral chain ganglia total RNA using the SuperScript Choice System (Invitrogen)
in structural, functional, electrophysiologic and immu- and an HPLC-purified oligo-dT primer containing an adjacent
nohistochemical characteristics (15–20). Therefore, the T7 RNA polymerase promoter (GENSET Corporation, La Jolla,
CA). Biotinylated cRNA was transcribed from cDNA templates
SCG acts more as a simple ‘‘relay station,’’ whereas pre-
using an Enzo BioArray High Yield Transcript Labeling Kit
vertebral ganglia such as the SMG-CG appear to mediate (Affymetrix, Inc., Santa Clara, CA). Biotinylated cRNA was
the complex integration of a variety of visceral reflexes. purified using RNeasy columns, fragmented and hybridized to
Despite improved understanding of the anatomy and rat genome U34A oligonucleotide arrays (Affymetrix) follow-
function of prevertebral and paravertebral ganglia, the ing the manufacturer’s recommended protocol. After 16 hours
biochemical differences underlying their distinct function hybridization, arrays were washed and stained with streptavi-
and the basis for the differences in their response to di- din-phycoerythrin. Hybridization signals were then amplified
abetes remain poorly understood. Furthermore, the mo- by labeling stained arrays with a biotinylated goat anti-strep-
lecular alterations that precede the development of NAD tavidin antibody (Vector Laboratories, Burlingame, CA) and
restaining with streptavidin-phycoerythrin (Affymetrix EUK-
and the time course with which these alterations occur
GE-WS2 version 4 protocol). Arrays were scanned twice at 570
have not previously been investigated. Therefore, to bet-
nm using an Affymetrix GeneArray scanner.
ter define molecular differences between sympathetic
ganglia and their selective vulnerability to diabetes-in- Analysis of Microarray Data
duced NAD, we have used oligonucleotide microarrays
Microarray image data was analyzed using Affymetrix Mi-
to compare the transcriptomes of adult rat SCG and croarray Analysis Suite 5.0 (MAS5). For each experiment, the
SMG-CG, both before and after the onset of STZ-induced MAS5 software was used to perform 3 relevant, binary array
diabetes. Data from this report demonstrates selective comparisons (i.e. control SCG vs control SMG-CG, control
gene expression alterations in SCG and SMG-CG ganglia SCG vs diabetic SCG, control SMG-CG vs diabetic SMG-CG).
prior to onset of NAD, which may represent the molec- The software output provided a statistical call regarding differ-
ular basis for the development of this neuropathology. ential hybridization signal between the 2 arrays (‘‘increase,’’
‘‘decrease,’’ ‘‘no change’’) and a calculated fold change in gene
MATERIALS AND METHODS expression. For the purposes of this study analysis, only probe
sets that showed a consistent ‘‘increase’’ or ‘‘decrease’’ in all
Animals and Surgical Procedures 3 independent experiments were considered. The complete
Male Sprague-Dawley rats (;300 grams; Charles Rivers, MIAME-compliant data set is available at http://bioinformatics.
Belmont, MA) received a single dose of STZ (65 mg/kg, i.v.; wustl.edu.
Upjohn, Kalamazoo, MI) in 0.01 M citrate/saline buffer (pH
4.5) and within a few days developed plasma glucose levels Real-Time Quantitative RT-PCR
$350 mg% (mg/dl). Animals were housed and cared for in Total RNA was isolated from ganglia using TRIzol reagent
accordance within the guidelines of the National Institutes of (Invitrogen) following the manufacturer’s recommendations.

J Neuropathol Exp Neurol, Vol 63, November, 2004


1146 CARROLL ET AL

One mg of each total RNA was reverse transcribed in a 20-ml were performed in parallel with each primer set to verify an
reaction containing random hexamer primers and Moloney mu- absence of contamination.
rine leukemia virus reverse transcriptase; a parallel reaction,
performed identically except for the addition of reverse tran- RESULTS
scriptase, was used to verify an absence of genomic DNA con-
Expression Profiling of Normal Rat SMG-CG and SCG
tamination. After completion of reverse transcription, samples
Sympathetic Ganglia
were diluted to 100 ml with distilled water, boiled for 5 min
and stored at 2808C until used in real-time PCR experiments. To test the hypothesis that the differing response of
Primers for real-time quantitative RT-PCR of b-actin and test prevertebral and paravertebral sympathetic ganglia to a
mRNA were designed using Primer Express software (Version variety of experimental and clinical insults reflects, in
2.0; Applied Biosystems, Inc., Foster City, CA) following the part, distinct transcriptional programs, we enumerated the
manufacturer’s recommendations. Real-time PCR primers were
transcriptome of normal SCG and SMG-CG in 3 inde-
designed from the following sequences: rat b-actin, GenBank
pendent experiments. We identified 141 unique tran-
accession # NM03144; rat prepronociceptin exon 2, GenBank
accession # X97374; rat VGF, GenBank accession # M74223; scripts that were consistently expressed at higher levels
rat chromogranin B, GenBank accession # AF019974; rat in- in SMG-CG relative to SCG, and 118 unique transcripts
sulin-like growth factor I (IGF-I), GenBank accession # that were expressed at lower levels (Table 1). Most no-
X06043. The sequences of the real-time PCR primers designed tably, we identified numerous differences in the expres-
from these sequences are as follows: sion of transcripts coding for specific neurotransmitters

Downloaded from http://jnen.oxfordjournals.org/ by guest on March 21, 2016


(e.g. somatostatin, prepronociceptin, and proenkephalin)
b-actin forward primer, TTCAACACCCCAGCCATGT
and neurotransmitter receptors (e.g. tachykinin receptor
b-actin reverse primer, GTGGTACGACCAGAGGCATACA
1, cholecystokinin A receptor, G protein-coupled receptor
Prepronociceptin forward primer, CCCTTATGTGTTTCAGCC
TCTTG 51 [forms part of the GABA-B receptor], serotonin re-
Prepronociceptin reverse primer, GCAGCAGGACATCACAA ceptor and solute carrier family 6, member 4 [a serotonin
AACAG transporter]). We also found major differences in expres-
VGF forward primer, CCCGTTGGTCATGAAAACCT sion of multiple genes encoding proteins necessary for
VGF reverse primer, CCCCCGGATGAGTAGAAGGA synaptic structure and synaptic vesicle transport, fusion,
Chromogranin B forward primer, CCAGTGGATAACAGGGA and membrane recovery (e.g. synaptic vesicle glycopro-
TCACA tein 2b, synaptic cell adhesion molecule [similar to
Chromogranin B reverse primer, TAGGGCATTTGAGAGGA RA175 OC 363058], Arg/Abl-interacting protein
CTTCAA ArgBP2). Given the cytoskeletal abnormalities evident in
IGF I forward primer, GCCACGTCACCGCAAGAT
NAD, it was also notable that a large number of cyto-
IGF I reverse primer, TGGCAGGTGTTCCGATGTT
skeletal protein transcripts (e.g. neurofilament heavy pep-
The relative abundance of each test transcript was determined tide, tubulin, peripherin, and actin) were expressed at
using an ABI 5700 Gene Quantitation System (Applied Bio- substantially lower levels in SMG-CG relative to SCG.
systems) and normalized to the levels of b-actin mRNA in the Thus, as predicted by anatomic and functional studies,
same specimen. Our microarray analyses showed no evidence major biochemical differences exist between these pre-
of alterations in b-actin mRNA levels in diabetic SMG-CG or vertebral and paravertebral sympathetic ganglia. Many of
SCG. In addition, preliminary experiments in which we assayed these differences involve transcripts encoding proteins
b-actin mRNA levels in control and diabetic SMG-CG and whose function is likely affected during the development
SCG by real-time PCR and normalized these values to the lev-
of NAD.
els of 18S ribosomal RNA in the same specimens showed no
evidence of alterations in b-actin mRNA levels, indicating that Identification of Genes with Altered Expression in
it was an appropriate standard for normalization. Amplification
Diabetic Sympathetic Ganglia
of each specimen was performed in 6 replicates of a 25-ml
reaction containing 1 ml of reverse transcription reaction and To identify genes whose expression is altered prior to
SYBR Green PCR Master Mix per the manufacturer’s recom- the overt development of the morphologic lesions char-
mendations (Applied Biosystems). Preliminary experiments acteristic of NAD, rats were injected with a single dose
were performed to determine the optimal concentration of each of streptozotocin, a treatment that produced diabetes as-
forward and reverse primer and to verify that each primer set sociated with markedly elevated plasma glucose levels
demonstrated similar amplification efficiencies under our reac-
(Table 2). The expression profiles of diabetic SCG and
tion conditions. The relative quantity of each test mRNA was
then established by normalizing the level of the corresponding
SMG-CG were compared with those of control SCG and
cDNA to that of b-actin cDNA in the same reverse transcription SMG-CG and consistent differences in all 3 experiments
reaction. Dissociation curves were performed after the comple- were identified (Table 3). While we identified 82 tran-
tion of each PCR reaction to verify an absence of extraneous scripts with altered expression in diabetic SMG-CG, us-
reaction products (e.g. primer-dimers) that might confound the ing the same criteria we identified a total of only 28 tran-
quantitation of each target. Controls lacking added template scripts whose expression was changed in diabetic SCG.

J Neuropathol Exp Neurol, Vol 63, November, 2004


GENE EXPRESSION IN DIABETIC GANGLIA 1147

In pre-NAD, diabetic SMG-CG, we observed upregu- potentially important in NAD pathogenesis, we quanti-
lation of transcripts encoding multiple neuropeptides tated the expression of selected mRNA in pre-lesion di-
(VGF nerve growth factor-inducible secreted protein, abetic ganglia and ganglia with NAD. For these experi-
prepronociceptin) and neurotransmitter receptors (puri- ments, cDNA was prepared from RNA isolated from
nergic receptor P2X2, serotonin receptor). The expression SMG-CG and SCG collected from rats 2 or 17 weeks
of mRNA encoding proteins essential for synapse struc- after the injection of streptozotocin or vehicle. Real-time
ture and function (N-ethylmaleimide sensitive factor quantitative PCR was then used to assay the levels of
(NSF), RAB 3A, RAB 11B, synapsin, chromogranin B, mRNA-encoding neuropeptides (VGF, prepronociceptin),
solute carrier family 18, membrane 1 [a chromaffin gran-
synaptic function (chromogranin B), or growth factors
ule amine transporter], G PCR kinase-interactor 1) was
(IGF-I). The assayed levels of these messenger RNAs
typically increased in diabetic SMG-CG, although the ex-
pression of a number of transcripts was significantly de- were normalized to the levels of b-actin mRNA in the
creased (monoamine oxidase A, B/K protein [synaptotag- same cDNA.
min], annexin V, similar to MUM-2 protein). Increased Using this approach, we found that each of the mRNAs
expression of cytochrome P450 and diabetes-inducible whose expression was altered specifically in prelesion di-
P450RLM6 (which may eventually prove to be the same abetic SMG-CG showed qualitatively similar changes by
gene), as well as genes encoding enzymes essential for real-time PCR (Fig. 1). We did note that the exact fold
energy metabolism (particularly glycolysis, oxidative change determined by real-time quantitative PCR differed

Downloaded from http://jnen.oxfordjournals.org/ by guest on March 21, 2016


phosphorylation, and lipid metabolism) were also iden- somewhat from the fold change determined in microarray
tified. On the other hand, diabetes was associated with a experiments, a common finding in this type of compari-
marked decrease in the expression of transcripts coding son that has been described by several other laboratories.
for mitochondrial uncoupling protein, as well as several These alterations in mRNA expression persisted to 17
other mitochondria-based enzymes. A transcript encoding weeks after the induction of diabetes, a time well after
galectin-related inter-fiber protein also demonstrated a the morphologic lesions of NAD have begun to appear.
striking and consistent 10-fold decrease in expression. In contrast, none of these transcripts demonstrated
In contrast to SMG-CG, we identified very few chang-
mRNA levels that were significantly altered in diabetic
es in the transcriptome of diabetic SCG. For example, we
SCG relative to SCG from vehicle-treated animals.
identified only 2 synapse-related transcripts (similar to
VAT-1, diazepam binding inhibitor) that were altered in DISCUSSION
diabetic SCG. Furthermore, diabetic SCG exhibited less
prominent increases in transcripts encoding enzymes in- Comparison of the Gene Expression Profiles of SCG
volved in glycolysis, oxidative phosphorylation, and lipid and SMG-CG in Control Rats
metabolism than were observed in prelesion diabetic
Neurons within normal prevertebral sympathetic gan-
SMG-CG.
Overall, diabetes-induced changes in gene expression glia differ from those in paravertebral chain ganglia in
were markedly different between sympathetic ganglia structural, functional, electrophysiologic, and immuno-
types. For example, expression of mRNA encoding ty- histochemical characteristics (15–20). Our systematic mi-
rosine hydroxylase, a key enzyme in norepinephrine syn- croarray analysis has also demonstrated marked differ-
thesis, is increased in diabetic rat SMG-CG but not in ences in gene expression between pre- and paravertebral
diabetic SCG, consistent with our previous studies indi- ganglia. Although we cannot exclude the possibility that
cating that the activity of this enzyme is selectively in- some differentially expressed mRNA is expressed by oth-
creased in diabetic SMG-CG (21). In fact, of the 110 er cell types within each ganglion (e.g. Schwann and sat-
differentially expressed transcripts that were identified ellite cells), it is likely that many of these transcripts are
(82 in SMG-CG and 28 in SCG), only 5 demonstrated neuronally derived. The differences we have identified in
consistent changes in both types of diabetic ganglia (Ta- the expression of somatostatin and prepronociceptin (both
ble 3). These include mRNA for mitochondrial uncou- increased selectively in prevertebral ganglia) and the ex-
pling protein (UCP-1), early growth response 1 (NGF- pression of serotonin receptors and transporters, calbin-
IA/Krox-24/EGR1), and cytochrome oxidase, all of din, G protein-coupled receptor 51 [GABA-B receptor]
which are substantially decreased in both ganglia, and
and proenkephalin (all selectively increased in the SCG)
‘‘upregulated by vitamin D1’’ (Vdup1) and diabetes-in-
parallel the results of immunohistochemical and in situ
ducible cytochrome P450RLM6, which are modestly in-
hybridization studies in rat and other species (16, 22–29)
creased in both ganglia.
and support the validity of our microarray analyses. Fur-
Confirmation of Diabetes-Responsive Gene Expression ther, the substantially higher expression of mRNA-encod-
in Sympathetic Ganglia ing tachykinin receptor 1 and cholecystokinin A receptor
To validate the results of our microarray analyses and in SMG-CG compared to SCG is consistent with the
to further examine the expression of a set of transcripts known collateral projections of DRG and possibly vagal

J Neuropathol Exp Neurol, Vol 63, November, 2004


TABLE 1 1148
Differential Gene Expression in Non-Diabetic Sympathetic SMG-CG versus SCG

SMG-CG specific expression (n 5 141) SCG specific expression (n 5 118)

Locus link Gene description Fold D Locus link Gene description Fold D

Synaptic structure/function Synaptic structure/function


24797 Somatostatin 35.2 29237 Preproenkephalin, related sequence 30.2
24889 Cholecystokinin A receptor 14.4 25553 Solute carrier family 6, member 4 14.9
24807 Tachykinin receptor 1 4.2 79246 5-hydroxytryptamine (serotonin) receptor 3a 7.2
25516 Prepronociceptin* 3.3 24166 Adenylate cyclase activating peptide I 4.4
114901 Arg/Abl-interacting protein ArgBP2 2.6 117556 Synaptic vesicle glycoprotein 2 b 4.3
24259 Chromogranin B* 2.5 116457 MAP kinase 8 interacting protein 3.0
29483 Solute carrier family 1, member 3 2.1 24166 Adenylate cyclase activating polypeptide 1 2.8
25380 Annexin 1 2.1 363058 Similar to RA175 OC363058 2.4

J Neuropathol Exp Neurol, Vol 63, November, 2004


83633 G protein-coupled receptor 51 2.2
Biosynthetic and metabolic pathways Biosynthetic and metabolic pathways
24340 Carboxyl esterase (ES-10) 21.3 81743 Cyclic GMP stimulated phosphodiesterase 7.0
362282 Phosphoenolpyruvate carboxykinase 1 11.2 25739 Brain glycogen phosphorylase 3.7
113902 Carboxylesterase 3 7.0 64157 Dimethylarginine dimethylaminohydrolase 1 2.7
24539 Lipoprotein lipase 7.0 83792 Stearoyl-Coenzyme A desaturase 2 2.4
29184 CD36 antigen 4.7
79451 Fatty acid binding protein 4 4.5
54249 Adipsin 4.2
25703 Retinol binding protein 4 3.6
25698 Arginosuccinate synthetase 3.3
54232 Carbonic anhydrase 3 3.0
CARROLL ET AL

29289 Xanthine dehydrogenase 2.9


58952 Plasma glutamate carboxypeptidase 2.7
29692 Phospholipase A2, group IIA 2.6
29516 Phosphodiesterase 3B 2.6
Mitochondrial and oxidative stress Mitochondrial and oxidative stress
24294 Cytochrome P450, subfamily 11B 6.7
25756 Carnitine palmitoyltransferase 1b 4.6
25288 Fatty acid Coenzyme A ligase, chain 2 2.8
113976 Fatty acid Coenzyme A ligase, chain 4 2.7
171341 Microsomal glutathione S-transferase 1 2.3
Growth factors, cytokines, hormones Growth factors, cytokines, hormones
24180 Angiotensin II receptor, type 1 (AT1A) 4.6 60663 Insulin receptor-related receptor 4.4
24482 Insulin-like growth factor 1* 3.2
24484 IGF binding protein 3 3.1
Signaling/ECM/other Signaling/ECM/other
83839 Calbindin 1 8.9 25216 Syndecan 1 5.0
117130 Galectin-related inter-fiber protein 4.4 84014 RalA binding protein 1 3.9
116745 K channel, subfamily H, member 6 4.2 25523 Protein kinase, cGMP- dependent, type II 3.4
24877 Visinin-like 1 3.2 24245 Calcium/calmodulin-dependent protein kinase 3.3
24330 Early growth response 1 3.4 25510 Purkinje cell protein 4 3.2

Downloaded from http://jnen.oxfordjournals.org/ by guest on March 21, 2016


GENE EXPRESSION IN DIABETIC GANGLIA 1149

only known genes with a greater than 2-fold expression difference are represented in this table. The full MIAME-compliant data set is available at http://bioinfor-
Locus Link ID, gene description, and average fold increase of gene expression in indicated ganglion are shown (n 5 3 independent experiments). For brevity,
TABLE 2
Serum Glucose Levels in Diabetic and Control Rats

Fold D

2.7
2.5
2.2
2.2
Duration N Glucose
Group of diabetes (rats) (mg% 6 SEM)

Diabetic 2 weeks 12 490 6 12


Control 12 109 6 31
Diabetic 2 weeks 12 454 6 14
Control 12 115 6 31
SCG specific expression (n 5 118)

Actinin alpha 2 associated LIM protein


6

Ptpn1 non-receptor phosphatase type 1


Diabetic 6 weeks 12 510 16
Control 11 114 6 4
Diabetic 17 weeks 4 422 6 21
Control 4 116 6 2
Gene description

Neurofilament heavy peptide

1
Glucose levels were assayed in a representative animal from
each cage (4 total).

sensory axons as they pass through the prevertebral gan-


glia to visceral targets (13–15).
matics.wustl.edu. Transcripts whose altered expression was confirmed by RT-PCR are indicated with an asterisk.

Downloaded from http://jnen.oxfordjournals.org/ by guest on March 21, 2016


The explanation that differences in neurotrophin sup-
Keratin 19

port are responsible for the variation in ganglionic re-


sponse between SCG and SMG-CG has been advanced
(30) and disputed (31). Our current studies are consistent
Signaling/ECM/other

with our previous immunohistochemical and in situ hy-


bridization studies (31) demonstrating that trkA and
p75NTR dominate the neurotrophin receptor profile in both
TABLE 1 (Continued)

Locus link

24587
360626
114108
24697

SMG-CG and SCG, far exceeding the expression of trkB


and trkC. IGF-I gene expression is 2.8-fold higher in nor-
mal SMG-CG compared to the SCG, which suggests the
possibility of autocrine or paracrine release of this im-
portant neurotrophic substance. Normal SMG-CG and
SCG also differ markedly in expression of multiple genes
Fold D

3.1
2.9
2.8
2.4

encoding proteins involved in synaptic function or struc-


ture, including elements important in vesicle transport,
fusion, and synaptic membrane recovery, which may un-
derlie differences in the development of synaptic pathol-
ogy and NAD.
SMG-CG specific expression (n 5 141)

Comparison of Diabetic and Control Rat Ganglia


Amiloride-sensitive cation channel 1

Experimental and clinical diabetes differentially target


Sodium channel, type 1, alpha

the SMG-CG and SCG and their peripheral projections


Gene description

in human (9) and several experimental rodent models (6,


32), including the STZ-diabetic rat. We found that dia-
betes alters the expression of multiple genes in both SCG
and SMG-CG. Remarkably, only 5 genes of a combined
total of 110 are increased in expression in both diabetic
Calbindin 2

SMG-CG and diabetic SCG relative to their respective


Lumican

control ganglia. This degree of ganglionic specificity in


diabetes-modulated gene expression is striking and un-
anticipated. STZ-diabetic rats develop NAD in postgan-
Signaling/ECM/other

glionic noradrenergic axons innervating the ileal intra-


mural ganglia but not adjacent, equally lengthy
noradrenergic axons innervating the mesenteric vascula-
Locus link

117059

ture (4), which may reflect the origin of these axons from
81574
25364
81682

pre- and paravertebral ganglia, respectively (7, 8). Selec-


tive involvement of the nitrergic innervation of the stom-
ach of NOD mice (33) and differences in the degenerative

J Neuropathol Exp Neurol, Vol 63, November, 2004


1150
TABLE 3
Altered Gene Expression in Diabetic Sympathetic Ganglia versus Control

Altered in diabetic SMG-CG (n 5 82) Altered in diabetic SCG (n 5 28)

Locus link Gene description Fold D Locus link Gene description Fold D

Synaptic structure/function Synaptic structure/function


25516 Prepronociceptin* 12.5 NA Similar to Vat-1 AA892520 11.3
114115 Purinergic receptor P2X 12.2 25045 Diazepam binding inhibitor 21.5
29461 VGF nerve growth factor inducible* 12.1
60355 N-ethylmaleimide sensitive factor 12.1
25693 Solute carrier family 18, member 1 11.9
25085 Tyrosine hydroxylase 11.8
24765 Secretogranin 2 11.7

J Neuropathol Exp Neurol, Vol 63, November, 2004


24259 Chromogranin B* 11.6
83709 G PCR kinase-interactor 1 11.6
266668 NMDA receptor glutamate-binding chain 11.5
79434 RAB11B 11.5
25531 RAB3A 11.4
29179 Synapsin 11.4
59104 Neurotransmitter-induced early gene 4 21.5
29253 Monoamine oxidase A 21.4
192189 B/K protein (synaptotagmin) 21.3
NA Similar to MUM-2 protein (LOC287427) 21.3
25673 Annexin 5 21.3
Biosynthetic and metabolic pathways Biosynthetic and metabolic pathways
CARROLL ET AL

NA AI176456- Metallothionein-like 15.0 29692 Phospholipase A2, group IIA 12.7


25058 Hexokinase 1 11.6 29184 CD36 antigen 12.4
117596 ATPase, beta 56/58 kDa, isoform 2 11.5
246074 Stearoyl-Coenzyme A desaturase 1 2 2.9
24539 Lipoprotein lipase 2 1.8
84050 Ectonucleotide phosphodiesterase 2 2 1.7
24679 Prkar2b 2 1.5
50555 UDP-glucuronosyltransferase 8 2 1.5
Mitochondrial and oxidative stress Mitochondrial and oxidative stress
25086 Cytochrome P450, subfamily 2E, polypep- 10.4
tide 1
NA S48325- diabetes-inducible P450RLM6 13.6 NA S48325- diabetes-inducible P450RLM6 11.8
64317 Glutathione peroxidase 3 12.0
24294 Cytochrome P450, subfamily 11B, polypep- 11.7
tide 2
NA D00636- NADH-cytochrome b5 reductase 11.4
24860 Uncoupling protein 1 229.1 24860 Uncoupling protein 1 224.8
25250 Cytochrome c oxidase subunit VIII-H 215.0 25250 Cytochrome c oxidase subunit VIII-H 28.5
25756 Carnitine palmitoyltransferase 1b 27.2
24158 Acetyl-coenzyme A dehydrogenase, medium 24.7
chain

Downloaded from http://jnen.oxfordjournals.org/ by guest on March 21, 2016


GENE EXPRESSION IN DIABETIC GANGLIA 1151

Locus Link ID (where available), gene description, and fold change of expression in the indicated diabetic ganglion are shown. Transcripts with similarly altered
expression in both ganglia types are indicated in bold. Transcripts whose altered expression was confirmed by RT-PCR are indicated with an asterisk. For brevity,
Fold D

11.8
22.7
22.3

only representative known genes are displayed in this table. The full MIAME-compliant data set is available at http://bioinformatics.wustl.edu.
Upregulated by Vitamin D 1 (Vdup1)
Altered in diabetic SCG (n 5 28)

Neurofilament heavy peptide


Gene description

Early growth response 1

Downloaded from http://jnen.oxfordjournals.org/ by guest on March 21, 2016


Signaling/ECM/other
Locus link

117514
24587
24330
TABLE 3 (Continued)

Fig. 1. Real-time PCR validation of altered expression of


selected transcripts in control, pre-lesion, and NAD diabetic
Fold D

superior mesenteric/celiac ganglia and superior cervical gan-


11.8
210.9
24.3

glia. Real-time quantitative RT-PCR was used to quantitate lev-


els of insulin-like growth factor I (IGF-I), chromogranin B, pre-
pronociceptin, and VGF nerve growth factor-inducible mRNAs
in RNA isolated from pooled (8 to 24 ganglia per pool) superior
mesenteric/celiac or superior cervical ganglia collected from
rats injected with vehicle or streptozotocin after 2 weeks (‘‘pre-
Altered in diabetic SMG-CG (n 5 82)

Upregulated by Vitamin D 1 (Vdup1)

lesion,’’ A–D) or 17 weeks (‘‘post-lesion,’’ E–G). The levels


of synapse-associated protein messenger RNA were normalized
to the levels of b-actin mRNA measured in the same specimens.
Galectin-related inter-fiber protein

The normalized levels of each mRNA in normal superior mes-


enteric/celiac ganglia were used as a reference (1-fold), with
Gene description

the mRNA levels in all other ganglia calibrated relative to this


Early growth response 1

standard (indicated by fold change on the vertical axis). Rela-


tive expression levels determined for each mRNA are repre-
sented by bars, with standard errors of the mean indicated for
each bar.

response of established dissociated SCG and CG neuron


cultures to a high glucose medium challenge in vitro (34)
have also been reported. Our demonstration that diabetic
Signaling/ECM/other

SMG-CG and SCG develop distinct patterns of gene ex-


pression prior to the development of the morphologic le-
sions of NAD may provide a molecular underpinning for
Locus link

the differential development of this pathology. In light of


117514
117130
24330

the selectivity of NAD for the SMG-CG, it is notable that


diabetes resulted in the altered expression of nearly 3-
fold more transcripts in the SMG-CG than SCG. Further,

J Neuropathol Exp Neurol, Vol 63, November, 2004


1152 CARROLL ET AL

the proteins encoded by these diabetes-responsive also regulated in vivo by electrical activity, injury, sei-
mRNAs mediate functions that provide important insights zures, and feeding and its expression is modulated in as-
into the molecular mechanisms resulting in NAD. sociation with circadian variation (44), synaptic remod-
eling, and axonal sprouting (45). Of particular relevance
Synaptic Structure/Function Genes to diabetes, VGF plays a critical role in energy balance,
NAD in the diabetic rat SMG-CG is a progressive, with mice with a targeted deletion of this gene becoming
synapse-directed process requiring 3 or more months to hypermetabolic and demonstrating markedly reduced lep-
develop as gauged by ultrastructural criteria. As NAD tin levels and fat stores (46, 47).
develops, dystrophic presynaptic axon terminals accu-
mulate neurotransmitter granules, clathrin-coated vesi- Mitochondria and Oxidative Stress
cles, tubulovesicular elements, disorganized microtu- Oxidative stress in diabetic peripheral nerve (48) is
bules, and neurofilaments. In the SCG, which does not thought to result from a variety of physiologic and path-
develop NAD, these abnormal accumulations are not ev- ophysiologic pathways (e.g. metabolism of nitric oxide,
ident. Synapse-directed processes, particularly those re- polyol pathway, arachidonic acid and catecholamines,
lated to synaptic vesicle function, membrane retrieval, leukocyte function, ischemia, formation of glycated pro-
and turnover of subcellular organelles, are thus likely key teins). Brownlee has proposed that a diabetes-induced in-
targets during the pathogenesis of NAD. crease in endothelial cell glucose metabolism produces

Downloaded from http://jnen.oxfordjournals.org/ by guest on March 21, 2016


We grouped genes involved in neurotransmitter syn- an exaggerated proton gradient in mitochondria, resulting
thesis and metabolism, receptor synthesis, and synapse- in the generation of excess ubiquinone intermediates and
directed structural and functional elements into a single increased superoxide production (49), although others
category for comparison. In the SMG-CG of diabetic an- have reported an insulin- and NT-3-sensitive decrease in
imals, the expression of 18 genes encoding proteins in- mitochondrial membrane potential in adult cultured DRG
volved in synaptic structure/function genes is altered. In neurons (50). Any of these processes may generate in-
contrast, only 2 genes showed minimally altered expres- creased amounts of reactive oxygen species (ROS) in the
sion in the SCG from the same diabetic animals and these sympathetic ganglia of diabetic animals, particularly in
were different transcripts. Abnormalities affecting vari- dystrophic nerve terminals, which may contain large ag-
ous nerve terminal constituents, particularly those in- gregates of mitochondria (32).
volved in neurotransmitter release and recycling, are In the SMG-CG, diabetes alters the expression of nu-
thought to play a role in the development of synaptic merous transcripts encoding mitochondrial proteins
pathology in neurodegenerative diseases (35). Our studies whose function is relevant to the metabolic alterations
show diabetes likewise results in a ganglion-specific noted above. Particularly striking is a 20- to 30-fold de-
change in many synaptically directed genes. Increased crease in the expression of uncoupling protein (UCP)-1,
expression of chromogranin B and solute carrier family a protein initially identified on the basis of its thermo-
18, member 1 (chromaffin granule amine transporter) genic role (51). Numerous recent studies have demon-
genes in diabetic SMG-CG may contribute directly to strated that UCPs are also involved in ATP synthesis,
abnormal neurotransmitter granule number, structure, or control of the NAD1/NADH ratio, weight regulation,
function, perhaps providing a surplus of the enlarged, dis- control of fatty acid metabolism, and flux of lipid sub-
organized or neurotransmitter-deficient nerve terminal strates across the mitochondrial membrane (52), and, in
synaptic vesicles and their membranous aggregates prom- particular, control of the generation of ROS. Overexpres-
inent in NAD. Diabetes also results in the SMG-CG-spe- sion of UCP in transgenic mice decreases the diabetes-
cific altered expression of N-ethylmaleimide sensitive induced proton gradient and normalizes deranged polyol,
factor (NSF), secretogranin 2, B/K protein (synaptotag- advanced glycosylation endproduct, protein kinase C, and
min), annexin V, several Rabs (Rab 3A, Rab 11B), syn- hexosamine pathways (53). Exposure to high glucose in
apsin and purinergic receptor P2X, which are known to vitro and in vivo also decreases expression of UCP-3, an
participate in synaptic vesicle transport, aggregation, event associated with increased generation of ROS (54).
docking, fusion, Ca12 dynamics, neurotransmitter release, Diabetes-induced reductions in ganglionic UCP-1 ex-
and reuptake (36–41). Also increased is G PCR kinase pression is thus likely to have multiple effects potentially
interactor 1 protein, a molecule known to regulate b-2 contributing to NAD pathogenesis.
adrenergic receptor endocytosis and, in excess, to hinder Of note, UCP-1 expression is decreased in both dia-
b2-adrenergic receptor signaling (42). VGF, a secreted betic SCG and SMG-CG and so cannot by itself be re-
protein whose expression is increased more than 2-fold sponsible for selectivity of neuroaxonal dystrophy for
in diabetic SMG-CG (but not SCG) is an NGF- and SMG-CG. However, the expression of cytochrome P450,
BDNF-inducible gene product that is widely distributed subfamily 2E (CYP2E1), which is present in both mito-
in the CNS and PNS where it undergoes regulated release chondrial and microsomal intracellular compartments, is
from dense core secretory vesicles (43). VGF release is increased more than 10-fold in the diabetic SMG-CG but

J Neuropathol Exp Neurol, Vol 63, November, 2004


GENE EXPRESSION IN DIABETIC GANGLIA 1153

is unchanged in the SCG. Increased expression of mito- have identified in our studies should be highly useful
chondrial CYP2E1 (55) has been proposed to increase markers for future investigations of the pathogenesis of
ROS production in diabetic rat brain. Further, expression this important diabetic autonomic neuropathy.
of diabetes-inducible cytochrome P450RLM6 (a gene
product that is highly similar and may prove to be iden- REFERENCES
tical to the CYP2E1 protein identified in diabetic rat liv- 1. Ewing DJ, Campbell IW, Clarke BF. The natural history of diabetic
er) is increased 1.8-fold in diabetic SCG and 3.6-fold in autonomic neuropathy. Quart J Med 1980;49:95–108
SMG-CG while expression of NADH cytochrome b5 re- 2. Rundles RW. Diabetic neuropathy: General review with report of
ductase, a superoxide generator (56) is increased 40% in 125 cases. Medicine (Baltimore) 1945;24:111–60
3. Hosking DJ, Bennett T, Hampton JR. Diabetic autonomic neurop-
the SMG-CG but is decreased 30% in the SCG of the athy. Diabetes 1978;27:1043–55
same animals. These alterations may interfere with re- 4. Schmidt RE. Neuropathology and pathogenesis of diabetic auto-
spiratory chain function in diabetic ganglia, resulting in nomic neuropathy. In: Tomlinson DR, ed. Neurobiology of diabetic
increased ROS production. Decreased expression of neuropathy. Amsterdam: Academic Press, 2002:267–92
UCP-1 may worsen mitochondrial dysfunction and, in 5. Schmidt RE. Neuronal preservation in the sympathetic ganglia of
rats with chronic streptozotocin-induced diabetes. Brain Res 2001;
combination with the respiratory chain abnormalities oc-
921:256–59
curring in SMG-CG, contribute to the pathogenesis of 6. Schmidt RE, Plurad SB. Ultrastructural and biochemical character-
NAD. ization of autonomic neuropathy in rats with chronic streptozotocin

Downloaded from http://jnen.oxfordjournals.org/ by guest on March 21, 2016


diabetes. J Neuropathol Exp Neurol 1986;45:525–44
Other Mechanisms 7. Hill CE, Hendry IA, Ngu MC, Van Helden DF. Subpopulations of
sympathetic neurons differ in their sensitivity to nerve growth fac-
There are modest increases in the expression of tran-
tor antiserum. Dev Brain Res 1985;23:121–30
scripts encoding several enzymes in the glycolytic path- 8. Hill CE. Selectivity in sympathetic innervation during development
way, including hexokinase (1.6-fold), aldolase (1.3-fold: and regeneration in the rat. Experientia 1985;41:857–62
data not shown), and glyceraldehyde-3-phosphate dehy- 9. Schmidt RE, Plurad SB, Parvin CA, Roth KA. The effects of dia-
drogenase (1.3-fold: data not shown), of diabetic SMG- betes and aging on human sympathetic autonomic ganglia. Am J
CG without comparable change in the SCG of the same Pathol 1993;143:143–53
10. Schmidt RE, Plurad SB, Modert CW. Neuroaxonal dystrophy in the
animals. Increased glycolytic flux would be expected to autonomic ganglia of aged rats. J Neuropathol Exp Neurol 1983;
increase mitochondrial activity, possibly resulting in an 42:376–90
increase in ROS production, and increase activity in com- 11. Schmidt RE, McAtee SJ, Plurad DA, Parvin CA, Cogswell BE,
plication-related polyol, glucosamine, protein kinase C, Roth KA. Differential susceptibility of prevertebral and paraverte-
and advanced glycosylation product pathways (49). bral sympathetic ganglia to experimental injury. Brain Res 1988;
460:214–26
The expression of the gene for galectin-related inter-
12. Kane WH, Johnson EM. Effect of chronic clonidine treatment and
fiber protein (GRIFIN), a key factor in injury-related withdrawal on tyrosine hydroxylase activity in peripheral ganglia
axon growth (57, 58), is 4.4-fold increased in normal rat and the locus ceruleus. Eur J Pharmacol 1978;51:309–12
SMG-CG compared to SCG, and responds to diabetes 13. Sejnowski TJ. Peptidergic synaptic transmission in sympathetic
with a 10-fold decrease in expression in the SMG-CG in ganglia. Fed Proc 1982;441:2923–28
14. Llewellyn-Smith IJ. Neuropeptides and the microcircuitry of the
the absence of an effect on the SCG. Decreased GRIFIN
enteric nervous system. Experientia 1987;43:813–21
expression in diabetic SMG-CG may interfere with ter- 15. Szurszewski JH. Physiology of mammalian prevertebral ganglia.
minal axon regeneration or collateral sprouting, thought Annu Rev Physiol 1981;43:53–68
to contribute to the pathogenesis of NAD (4). Galectin- 16. Elfvin LG, Lindh B, Hokfelt T. The chemical neuroanatomy of
3 upregulation in diabetic kidney protects against AGE- sympathetic ganglia. Annu Rev Neurosci 1993;16:471–507
induced tissue injury (59) and a similar role in sympa- 17. Janig W, McLachlan EM. Characteristics of function specific path-
ways in the sympathetic nervous system. Trends Neurosci 1992;15:
thetic ganglia could worsen AGE effects. 475–81
In summary, our analyses of the transcriptomes of the 18. Boyd HD, McLachlan EM, Keast JR, Inokuchi H. Three electro-
SMG-CG and SCG demonstrate that these ganglia ex- physiologic classes of guinea pig sympathetic postganglionic neu-
press remarkably distinct profiles of mRNA, consistent rons have distinct morphologies. J Comp Neurol 1996;369:372–87
with their distinct functions and differential susceptibility 19. Chau YP, Lu KS. Investigation of the blood-ganglion barrier prop-
erties in rat sympathetic ganglia by using lanthanum ion and horse-
to the development of NAD. Our findings also indicate
radish peroxidase as tracers. Acta Anat 1995;153:135–44
that the strikingly different responses of SMG-CG and 20. Wang HS, McKinnon D. Potassium channel expression in prever-
SCG to diabetes occurs before the development of the tebral and paravertebral sympathetic neurons: Control of firing
morphologic lesions of NAD and involves genes encod- properties. J Physiol (Lond) 1995;485:319–35
ing proteins mediating synaptic structure and function, 21. Schmidt RE, Cogswell BE. Tyrosine hydroxylase activity in sym-
pathetic nervous system of rats with streptozocin-induced diabetes.
mitochondrial function and responses to oxidative stress,
Diabetes 1989;38:959–68
glycolysis, and axon outgrowth. These observations sug- 22. Grkovic I, Anderson CR. Calbindin D28K-immunoreactivity iden-
gest that alterations in one or more of these functions are tifies distinct subpopulations of sympathetic pre- and postganglionic
important for the development of NAD. The genes we neurons in the rat. J Comp Neurol 1997;386:245–59

J Neuropathol Exp Neurol, Vol 63, November, 2004


1154 CARROLL ET AL

23. Mantyh PW, Catton MD, Allen CJ, Labenski ME, Maggio JE, Vig- ADP ribosylation factor GTPase-activating protein. Proc Natl Acad
na SR. Receptor binding sites for cholecystokinin, galanin, somato- Sci USA 1998;95:14082–87
statin, substance P and vasoactive intestinal polypeptide in sym- 43. Levi A. A protein induced by NGF in PC12 cells is stored in se-
pathetic ganglia. Neurosci 1992;46:739–54 cretory vesicles and released through the regulated pathway. EMBO
24. Anderson RL, Jobling P, Matthew SE, Gibbins IL. Development of J 1989;8:2217–23
convergent synaptic inputs to subpopulations of sympathetic neu- 44. Salton SR. Neurotrophins, growth factor regulated genes and the
rons. J Comp Neurol 2002;447:218–33 control of energy balance. Mt Sinai J Med 2003;70:93–100
25. Kummer W, Fischer A. Nociceptin and its receptor in guinea-pig 45. Snyder SE, Cheng HW, Murray KD, Isackson PJ, McNeill TH,
sympathetic ganglia. Neurosci Lett 1997;234:35–38 Salton SR. The messenger RNA encoding VGF, a neuronal peptide
26. Karhula T. Comparison of immunohistochemical localization of precursor, is rapidly regulated in the rat central nervous system by
[Met5] enkephalin-Arg6-Gly7-Leu8, [Met5] enkephalin, neuropep- neuronal activity, seizure and lesion. Neurosci 1998;82:7–19
tide Y and vasoactive intestinal polypeptide in the superior cervical 46. Hahm S, Tooru M, Mizuno T, et al. Targeted deletion of the Vgf
ganglion of the rat. J Auton Nerv Syst 1995;51:9–18 gene indicates that the encoded secretory peptide precursor plays a
27. Nishimura M, Sato K, Shimada S, Tohyama M. Expression of nor- novel role in the regulation of energy balance. Neuron 1999;23:
epinephrine and serotonin transporter mRNAs in the rat superior 537–48
cervical ganglion. Mol Brain Res 1999;67:82–86 47. Hahm S, Fekete C, Toonu MM, et al. VGF is required for obesity
28. Filipov AK, Couve A, Pangalos MN, Walsh FS, Brown DA, Moss induced by diet, gold thioglucose treatment and agouti and is dif-
SJ. Heteromeric assembly of GABA(B)R1 and GABA(B)R2 recep- ferentially regulated in pro-opiomelanocortin- and neuropeptide Y-
tor subunits inhibits Ca(21) current in sympathetic neurons. J Neu- containing arcuate neurons in response to fasting. J Neurosci 2002;
rosci 2000;20:2867–74 22:6929–38
29. Kaprielian Z, Cho KO, Hadjiargyrou M, Patterson PH. CD9, a ma- 48. Obrosova IG. How does glucose generate oxidative stress in pe-

Downloaded from http://jnen.oxfordjournals.org/ by guest on March 21, 2016


jor platelet cell surface glycoprotein, is a ROCA antigen and is ripheral nerve? In: Tomlinson DR, ed. Neurobiology of diabetic
expressed in the nervous system. J Neurosci 1995;15:562–73 neuropathy. Amsterdam: Academic Press, 2002:3–35
30. Dixon JE, McKinnon D. Expression of the trk gene family of neu- 49. Brownlee M. Biochemistry and molecular cell biology of diabetic
rotrophin receptors in prevertebral sympathetic ganglia. Devel complications. Nature 2001;414:813–20
Brain Res 1994;77:177–82 50. Fernyhough P, Huang T-J, Verkhratsky A. Mechanism of mito-
31. Schmidt RE, Dorsey DA, Selznick LA, et al. Neurotrophin sensi- chondrial dysfunction in diabetic sensory neuropathy. J Periph Nerv
tivity of prevertebral and paravertebral rat sympathetic autonomic Syst 2003;8:227–35
ganglia. J Neuropathol Exp Neurol 1998;57:158–67 51. Boss O, Muzzin P, Giacobino JP. The uncoupling proteins, a review.
32. Schmidt RE, Dorsey DA, Beaudet LN, et al. Non-obese diabetic Eur J Endocrinol 1998;139:1–9
(NOD) mice rapidly develop dramatic sympathetic neuritic dystro- 52. Ricquier D, Bouillaud F. The uncoupling protein homologues:
phy—A new experimental model of diabetic autonomic neuropathy. UCP1, UCP2, UCP3, StUCP and AtUCP. Biochem J 2000;345:
Am J Pathol 2003;163:2077–91 161–79
33. Cellek S, Foxwell NA, Moncada S. Two phases of nitrergic neu- 53. Vincent AM, Olzmann JA, Brownlee M, Sivitz WI, Russell JW.
ropathy in streptozotocin-induced diabetic rats. Diabetes 2003;52: Uncoupling proteins prevent glucose-induced neuronal oxidative
2353–62 stress and programmed cell death. Diabetes 2004;53:726–34
34. Lincoln J, Semra Y, Smith N. Effects of high glucose in vitro on 54. Vincent AM, Berent AR, Brownlee M, Russell JW. Uncoupling
adult sympathetic neurons from rat superior cervical and celiac gan- proteins are associated with regulation of high glucose induced ox-
glia. J Periph Nerv Syst 2003;8:188 idative stress and neuronal death [abstract]. J Periph Nerv Syst
35. LoPachin RM, Ross JF, Lehning EJ. Nerve terminals as the primary 2001;6:188
site of acrylamide action: A hypothesis. Neurotoxicol 2002;23:43–59 55. Raza H, Prabu SK, Robin M-A, Avadhani NG. Elevated mitochon-
36. Schiavo G, Stenbeck G. Molecular analysis of neurotransmitter re- drial cytochrome P450 2E1 and glutathione S-transferase A4-4 in
lease. Essays Biochem 1997;32:30–41 streptozotocin-induced diabetic rats. Diabetes 2004;53:185–94
37. Gotow T, Sakata M, Funakoshi T, Uchiyama Y. Preferential locali- 56. Whatley SA, Curti D, Das Gupta F, et al. Superoxide, neuroleptics
zation of annexin V to the axon terminal. Neurosci 1996;75:507–21 and the ubiquinone and cytochrome b5 reductases in brain and lym-
38. Gerdes H-H, Rosa P, Phillips E, et al. The primary structure of phocytes from normals and schizophrenic patients. Molecular Psy-
human secretogranin II, a widespread tyrosine-sulfated secretory chiatry 1998;3:227–37
granule protein that exhibits low pH-and calcium-induced aggre- 57. Sango K, Tokashiki A, Ajiki K, et al. Synthesis, localization and
gation. J Biol Chem 1989;264:12009–15 externalization of galectin-1 in mature dorsal root ganglion neurons
39. Geppert M, Goda Y, Hammer RE, et al. Synaptotagmin I: A major and Schwann cells. Eur J Neurosci 2004;19:55–64
58. Fukaya K, Hasegawa M, Mashitani T, et al. Oxidized galectin-1
Ca21 sensor for transmitter release at a central synapse. Cell 1994;
stimulates the migration of Schwann cells from both proximal and
79:717–27
distal stumps of transected nerves and promotes axonal regenera-
40. Li L, Chin L-S. The molecular machinery of synaptic vesicle exo-
tion after peripheral nerve injury. J Neuropathol Exp Neurol 2003;
cytosis. Cell Mol Life Sci 2003;60:942–60
62:162–72
41. Sperlagh B, Erdelyi F, Szabo G, Vizi ES. Local regulation of [(3)]-
59. Iacobini C, Amadio L, Oddi G, et al. Role of galectin-3 in diabetic
noradrenaline release from the isolated guinea-pig right atrium by
nephropathy. J Am Soc Nephrol 2003;14:S264–S270
P(2X)-receptors located on axon terminals. Brit J Pharmacol 2000;
131:1775–83 Received May 26, 2004
42. Premont RT, Claing A, Vitale N, et al. b2-adrenergic receptor reg- Revision received July 12, 2004
ulation by GIT1, a G protein-coupled receptor kinase-associated Accepted July 20, 2004

J Neuropathol Exp Neurol, Vol 63, November, 2004

You might also like