Journal of Neurochemistry - 2002 - Karten - Cholesterol accumulates in cell bodies but is decreased in distal axons of

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Journal of Neurochemistry, 2002, 83, 1154–1163

Cholesterol accumulates in cell bodies, but is decreased in distal


axons, of Niemann–Pick C1-deficient neurons

Barbara Karten,*, ,1
Dennis E. Vance,*,à,2,3 Robert B. Campenot§,2 and Jean E. Vance*,
*Canadian Institutes of Health Research Group on the Molecular and Cell Biology of Lipids and Departments of Medicine,
àBiochemistry and §Cell Biology, University of Alberta, Edmonton, Alberta, Canada

Abstract however, the cholesterol content of NPC1-deficient and


Niemann–Pick type-C (NPC) disease is characterized by a wild-type neurons as a whole was the same. This apparent
progressive loss of neurons and an accumulation of uneste- paradox was resolved when the cholesterol content of NPC1-
rified cholesterol within the endocytic pathway. Unlike other deficient distal axons was found to be less than of wild-type
tissues, however, NPC1-deficient brains do not accumulate axons. Cholesterol sequestration in cell bodies did not depend
cholesterol but whether or not NPC1-deficient neurons accu- on exogenously supplied cholesterol since the cholesterol
mulate cholesterol is not clear. Therefore, as most studies on accumulated before birth and did not disperse when neurons
cholesterol homeostasis in NPC1-deficient cells have been were cultured without exogenous cholesterol. The altered
performed in fibroblasts we have investigated cholesterol cholesterol distribution between cell bodies and axons
homeostasis in cultured murine sympathetic neurons lacking suggests that transport of cholesterol, particularly that
functional NPC1. These neurons did not display obvious synthesized endogenously, from cell bodies to distal axons is
abnormalities in growth or morphology and appeared to impaired in NPC1-deficient neurons.
respond normally to nerve growth factor. Filipin staining Keywords: axons, cholesterol, filipin, neurons, Niemann–
revealed numerous cholesterol-filled endosomes/lysosomes Pick C, U18666A.
in NPC1-deficient neurons and the mass of cholesterol in cell J. Neurochem. (2002) 83, 1154–1163.
bodies was greater than in wild-type neurons. Surprisingly,

Niemann–Pick type-C (NPC1) disease is a fatal, autosomal et al. 1989) in which unesterified cholesterol accumulates in
recessive disorder that leads to premature death (Pentchev late endosomes/lysosomes. In these cells, the transport of low
et al. 1995; Vanier and Suzuki 1998). The disease is caused density lipoprotein (LDL)-derived cholesterol from late
in 95% of cases by a mutation in the NPC1 gene resulting in endosomes/lysosomes to the plasma membrane is defective;
a lack of function of the NPC1 protein (Carstea et al. 1997).
The remaining 5% of NPC patients have a defect in the HE1
gene (Naureckiene et al. 2000). Patients with NPC disease Received July 5, 2002; revised manuscript received August 29, 2002;
accepted September 6, 2002.
exhibit increasing loss of motor control, seizures and other Address correspondence and reprint requests to Dr Jean E. Vance, 332
neuropathological symptoms (Pentchev et al. 1995; Vanier Heritage Medical Research Center, University of Alberta, Edmonton
1999). Similar to lysosomal storage diseases, NPC disease is Alberta T6G 2S2, Canada. E-mail: jean.vance@ualberta.ca
1
associated with axonal abnormalities (spheroids, meganeur- Supported by postdoctoral fellowships from the Alberta Heritage
ites and axonal dystrophy), demyelination, and formation of Foundation for Medical Research and the Deutsche Forschungsge-
meinschaft (Forschungsstipendium KA1578/1).
neurofibrillary tangles. With the exception of neurofibrillary 2
Medical Scientists of the Alberta Heritage Foundation for Medical
tangle formation, similar changes are observed in a murine Research.
3
model for NPC disease (Higashi et al. 1993; German et al. Holder of the Canada Research Chair in Molecular and Cell Biology of
2001). Lipids.
The exact function of the NPC1 protein is still unknown. Abbreviations used: HDL, high density lipoprotein; LAMP1, lyso-
some-associated membrane protein 1; LDL, low-density lipoprotein;
Essentially all biochemical studies on NPC1-deficient cells NGF, nerve growth factor; NPC, Niemann–Pick type C; PBS, phosphate-
have been performed in non-neuronal cells such as fibro- buffered saline; SDS, sodium dodecyl sulfate; U18666A, 3-b-(2-diethyl-
blasts and mutant CHO cells (Cadigan et al. 1990; Liscum aminoethoxy)androst-5-ene-17-one.

1154 Ó 2002 International Society for Neurochemistry, Journal of Neurochemistry, 83, 1154–1163
14714159, 2002, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1046/j.1471-4159.2002.01220.x by University Of British Columbia, Wiley Online Library on [01/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Cholesterol accumulation in npc–/– neurons 1155

it now also appears that the trafficking of endogenously acids 1261–1272 of human NPC1. A rabbit anti-mouse polyclonal
synthesized cholesterol is impaired (Lange and Steck 1998; antibody raised against amino acids 1254–1273 of murine NPC1
Cruz and Chang 2000; Lange et al. 2000). was provided by Dr W. S. Garver (University of Arizona, Tucson,
In the brain, the majority of cholesterol is derived from AZ, USA) and was used for the immunocytochemical studies. The
rat-anti mouse LAMP1 antibody was from PharMingen (Lexing-
endogenous synthesis since plasma lipoproteins do not
ton, KY). Alexa Fluor 488-labeled goat anti-rabbit IgG and Texas
cross the blood–brain barrier (Danik et al. 1999; Turley
Red-labeled goat anti-rat IgG were from Molecular Probes
et al. 1996; Turley et al. 1998). However, the increased (Eugene, OR, USA). U18666A [3-b-(2-diethylaminoethoxy)andr-
need for cholesterol during nerve regeneration is believed to ost-5-en-17-one] was purchased from Biomol Research Laborator-
be met by receptor-mediated uptake of apo E-containing ies (Plymouth Meeting, PA, USA). Other materials for gel
lipoproteins secreted by endoneurial macrophages which electrophoresis and immunoblotting were from Bio-Rad (Mississ-
salvage cholesterol released by degenerating neurons and auga, ON, Canada). Monoclonal antib-tubulin antibody (T 4026),
myelin (Goodrum et al. 2000). In NPC1-deficient mice, this filipin complex, and cholesterol were purchased from Sigma (St.
recycling mechanism for cholesterol has been reported to be Louis, MO, USA). Mouse 2.5S nerve growth factor was from
impaired (Goodrum and Pentchev 1997; German et al. Alomone Laboratories Ltd. (Jerusalem, Israel). Rat serum was
2002). In most tissues of NPC patients, cholesterol provided by the University of Alberta Animal Services. Delipi-
dated rat serum was prepared by the method of Cham and
accumulates but the relationship between this accumulation
Knowles (Cham and Knowles 1976). All other materials were
and the neurological manifestations is not understood. In
from Sigma, unless stated otherwise.
addition to cholesterol, glycosphingolipids also accumulate
in lysosomes/late endosomes of NPC1-deficient cells (Tan- Primary cultures of sympathetic neurons
iguchi et al. 2001; Zervas et al. 2001) and the concentration from NPC1-deficient mice
of glycosphingolipids in NPC brains is increased (Vanier A breeding colony of heterozygous Balb/cNctr-npcN/+mice, ori-
1999). In contrast, in the brains of NPC patients the ginally from the Jackson Laboratories (Bar Harbor, ME, USA), was
cholesterol content decreases with age (Vanier 1999; Xie established at the University of Alberta. Mice were maintained
et al. 1999). Thus, it has been suggested that NPC1- under temperature-controlled conditions with a 12-h light : 12-h
deficient neurons do not accumulate cholesterol (Elleder dark cycle. Hereafter, mice homozygous or heterozygous for the
et al. 1985). However, Dietschy and coworkers have mutation in the npc1 gene will be referred to as npc–/– or npc+/–,
respectively, whereas wild-type mice will be designated as npc+/+.
proposed that the decrease in cholesterol in NPC1-deficient
The genotype was determined from genomic DNA extracted from
brains is the result of an extensive loss of myelin which is
tail clippings of 1-day-old mouse pups in a PCR reaction as
rich in cholesterol (Xie et al. 2000), and that cholesterol described (Loftus et al. 1997). All experiments were performed by
does accumulate in neurons and glial cells. On the other comparing littermates of the three genotypes and were approved by
hand, Henderson et al. (2000) have found that the choles- the Health Sciences Animal Welfare Committee of the University of
terol content of cultured embryonic striatal neurons from Alberta. Superior cervical ganglia were dissected from one-day-old-
NPC1-deficient mice is normal. mouse pups. Ganglia of each mouse pup were placed separately into
In light of the conflicting and incomplete data on whether sterile microfuge tubes containing L15 medium with 10% rat serum
or not cholesterol accumulates in neurons lacking functional and 50 ng/mL NGF and maintained at 4°C overnight, but for no
NPC1, and since NPC disease is primarily a neurological longer than 24 h, prior to dissociation. This time period allowed
disorder, we have examined axonal growth and cholesterol genotyping of tail clippings from each mouse pup to be performed
and consequently ganglia of one genotype could be pooled prior to
homeostasis in primary neurons from NPC1-deficient mice.
dissociation. Neurons were plated either on 48-well plates at a
We show that NPC1 deficiency results in an accumulation of
density of 1 ganglion/well or into the center slot of compartmented
cholesterol in cell bodies but a reduction in the cholesterol culture dishes at a density of 0.6 ganglia/dish (Karten et al. 2002).
content of distal axons suggesting that the transport of Compartmented cultures were assembled as described previously
cholesterol from cell bodies into distal axons is impaired in (Campenot 1979; Campenot 1992). Briefly, a Teflon divider (Tyler
NPC1-deficient neurons. Research Instruments, Edmonton, AB, Canada) was sealed with
silicon grease to a 35-mm collagen-coated dish thereby partitioning
the dish into three compartments. Dissociated neurons were plated
Materials and methods in the center compartment. Within 2–3 days axons elongated along
the tracks, crossing under silicon grease barriers into left- and right-
Materials side compartments. Thus, the center compartment contained cell
Cell culture media and B27 supplement were purchased from bodies and proximal axons whereas side compartments contained
Invitrogen (Burlington, ON, Canada). Other cell culture materials pure distal axons. No exchange of fluid occurs between compart-
were from BD Biosciences (Bedford, MA, USA). The rabbit anti- ments (Campenot 1979). L15 medium with penicillin/streptomycin,
human polyclonal NPC1 antibody used for immunoblotting was a L-glutamine, glucose and the additives prescribed by Hawrot and

generous gift from Dr D. Ory (Washington University, St. Louis, Patterson (1979), including bicarbonate and methylcellulose, was
MO, USA) and was raised against a peptide consisting of amino used as basal medium to which was added 2.5% rat serum for the

Ó 2002 International Society for Neurochemistry, Journal of Neurochemistry, 83, 1154–1163


14714159, 2002, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1046/j.1471-4159.2002.01220.x by University Of British Columbia, Wiley Online Library on [01/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1156 B. Karten et al.

center compartment and 100 ng/mL NGF for the side compart- 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyltetrazolium bromide
ments. During the first week after plating, 10 lM cytosine (MTT tetrazolium salt) into an insoluble formazan product using
arabinoside and 20 ng/mL NGF were present in the center the CellTiter 96Ò Non-Radioactive Cell Proliferation Assay
compartment. Neurons were cultured for 10–12 days prior to the (Promega, Madison, WI, USA).
start of all experiments. In some experiments, neurons were placed
in serum-free medium consisting of a 1 : 1 mixture of Dulbecco’s Immunocytochemistry and filipin staining
modified Eagle’s medium and Ham’s F10 medium containing 0.4% Neurons in compartmented dishes or on collagen-coated coverslips
methylcellulose (w/v) with the same antibiotics and additives except were fixed for 20 min at room temperature in 4% paraformaldehyde,
that a B27 supplement mixture was used instead of serum. then blocked in 10% goat serum in phosphate-buffered saline
(PBS) ± 50 lg/mL filipin for 2 h at room temperature. For
Measurement of axonal extension immunocytochemical localization of LAMP1 and NPC1 the primary
Axonal extension was measured in compartmented dishes after antibodies used were rat anti-mouse LAMP1 (dilution 1 : 800) and
removal of distal axons (a process hereafter termed ÔaxotomyÕ) from rabbit anti-mouse NPC1 (dilution 1 : 500), respectively. Secondary
the side compartments with a jet of sterile distilled water (Campenot antibodies were Texas Red-labeled goat anti-rat IgG (dilution
1992). The wash was repeated twice to remove all traces of distal 1 : 200 for LAMP1) and Alexa Fluor 488-labeled goat anti-rabbit
axons from the side compartments without disturbing the collagen. IgG (dilution 1 : 200 for NPC1). Pictures were taken using a Zeiss
Axonal extension was measured using a Nikon Diaphot inverted LSM 510 confocal laser scanning microscope (Jena, Germany).
microscope equipped with a MD2 microscope digitizer (Accustage Excitation wavelengths were 351 nm (filipin), 488 nm (Alexa Fluor
Corp., Minneapolis, MN, USA) which tracks stage movements to an 488), and 543 nm (Texas Red).
accuracy of ± 0.005 mm. In each culture, 16 tracks were measured
in each side compartment. As an independent indicator of axonal Cholesterol biosynthesis
growth, the amount of axonal b-tubulin in distal axons was assessed Neurons were cultured in 48-well plates for 12 days then incubated
by immunoblotting. Proteins from distal axons were resolved on for 24 h with 0.4 lCi/mL [14C]acetate in basal medium containing
10% polyacrylamide gels containing 0.1% SDS under reducing 2.5% rat serum and 100 ng/mL NGF. Lipids were extracted with
conditions, then transferred to a nitrocellulose membrane and hexane/isopropanol 3 : 2 (v/v) and cholesterol was separated by
probed with anti-b-tubulin antibody (dilution 1 : 1000). Immuno- thin-layer chromatography in the solvent system hexane/diisopropyl
blots were developed using ECL reagent (SuperSignal West Dura ether/acetic acid 65 : 35 : 4 (v/v/v).
Extended, Pierce, Rockford, IL, USA).
Determination of cellular cholesterol content
Immunoblotting of NPC1 protein Cellular lipids were extracted twice with 2.5 mL hexane/isopropanol
Neurons were harvested into ice-cold homogenization buffer consist- 3 : 2 (v/v) after which an internal standard, 5a-cholestane, was
ing of 20 mM Tris-HCl, 1 mM EDTA, 0.25 mM sucrose (pH 7.4) with added. Unesterified cholesterol was converted to its trimethylsilyl
1 mM phenylmethylsulfonyl fluoride and a protease inhibitor cocktail ether derivative by reaction with bis(trimethylsilyl)trifluoroaceta-
(Complete Mini, Roche Diagnostics, Mannheim, Germany). Samples mide containing 1% trimethylchlorosilane in acetone for 15 min at
were homogenized in a glass homogenizer and nuclei and unbroken 50°C. The trimethylsilyl derivative was analyzed by gas–liquid
cells were removed from the homogenate by centrifugation at 1500 g chromatography.
for 10 min at 4°C. The supernatant was centrifuged in a TLA100.2
rotor (Beckman Instruments, Mississauga, ON, Canada) at 436 000 g Other methods
for 30 min at 4°C. The membrane pellet was resuspended in buffer Human lipoproteins were isolated by ultracentrifugation of plasma
containing 50 mM Tris/HCl (pH 7.4), 0.15 M NaCl, 2 mM EDTA and of normolipidemic volunteers in a KBr gradient (Sattler et al. 1994).
1% Triton X-100. Proteins were resolved on 6% polyacrylamide gels Concentrations of lipoproteins are given as mg protein/mL. Cellular
containing 0.1% sodium dodecyl sulfate (SDS) then transferred to protein was determined using the BCA Protein Assay (Pierce,
nitrocellulose and immunoblotted using anti-human NPC1 antibody Rockford, IL, USA).
(dilution 1 : 1000) and peroxidase-conjugated goat anti-rabbit IgG
(dilution 1 : 10 000) as secondary antibody. Immunoreactivity was
detected with ECL reagent (ECLWestern Blotting System, Amersham Results
Biosciences, Piscataway, NJ, USA).
Sympathetic neurons express NPC1 protein
NGF deprivation and neuronal survival
Even though npc–/– mice have no apparent phenotype at the
Neurons in 96-well plates were washed three times with L15
medium containing 0.4% methylcellulose with a 10-min incubation time of birth, only 12% of offspring generated by crossing
at 37°C between washes to ensure diffusion of the unstirred water npc+/– mice were homozygous for the npc1 mutation,
layer above the cells. Remaining traces of NGF were removed by indicating a prenatal lethality caused by the npc1 mutation.
incubation for 30 min with 100 lL medium containing 12 nM anti- This embryonic lethality apparently occurs after embryonic
NGF antibody (Cedarlane Laboratories Ltd, Hornby, ON, Canada). day 16 since Henderson and coworkers have reported that the
The cells were washed, then medium containing the indicated expected 25% of pups taken at this time are of the npc–/–
concentrations of NGF was added for 40 h. Neuronal survival genotype (Henderson et al. 2000).
was assessed based on the ability of the neurons to convert

Ó 2002 International Society for Neurochemistry, Journal of Neurochemistry, 83, 1154–1163


14714159, 2002, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1046/j.1471-4159.2002.01220.x by University Of British Columbia, Wiley Online Library on [01/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Cholesterol accumulation in npc–/– neurons 1157

Fig. 1 NPC1 is expressed in sympathetic neurons. Membrane-


enriched fractions were prepared from rat sympathetic neurons
maintained for 12 days in 24-well plates (lane 1: SCG), and from
homogenates of brain (lanes 2–4) and liver (lanes 5–7) from 5-week-
old npc+/+, npc+/– and npc–/– mice. Proteins were resolved by
electrophoresis on polyacrylamide gels and immunoblotted with anti-
human NPC1 antibody. Ten micrograms of protein were applied to
each lane.

NPC1 mRNA has been detected in several types of


neurons in the brain using in situ hybridization techniques
(Prasad et al. 2000) but, to our knowledge, the expression Fig. 2 Axonal extension is not dependent on NPC1 protein. Murine
sympathetic neurons were axotomized then axons were allowed to
of NPC1 protein in neurons has not been reported. We
regenerate in medium containing delipidated serum. (a) Axonal
therefore confirmed that sympathetic neurons express NPC1
extension. s, npc+/+; h, npc+/–; n, npc–/–. Data represent means ± SE
protein by immunoblotting using a polyclonal anti-human of four independent experiments with duplicate dishes and 30 tracks
NPC1 antibody. As shown in Fig. 1, lane 1, an immuno- measured per dish. (b) On day 7 after axotomy, distal axons were
reactive protein of 170 kDa, presumed to be NPC1, harvested and proteins analyzed by polyacrylamide gel electro-
comigrated with a protein from brains and livers of wild- phoresis and immunoblotting with antib-tubulin antibody. Each lane
type mice (Fig. 1, lanes 2 and 5). This protein was less contains protein from distal axons of 1 dish of neurons of each
abundant in brain and liver homogenates from npc+/– mice genotype. One representative experiment of four similar experiments
(Fig. 1, lanes 3 and 6), and was not detected in npc–/– mice is displayed.
(Fig. 1, lanes 4 and 7).

Neurons from npc–/– mice exhibit normal morphology genotypes we demonstrated by immunoblotting that the
and growth amount of b-tubulin, the most abundant cytoskeletal protein
To investigate if axonal growth of neurons is defective in of axons, was the same in distal axons of npc+/+, npc+/–
the absence of NPC1, we established conditions for culture and npc–/– neurons (Fig. 2b).
of primary sympathetic neurons from the superior cervical
ganglia of one-day-old npc–/–, npc+/– and npc+/+ mice in Survival of npc–/– neurons is not impaired
three-compartment dishes (Karten et al. 2002) by modifying in low concentrations of NGF
techniques previously used for rat sympathetic neurons Sympathetic neurons depend on NGF for survival and
(Campenot 1979). In this culture system, cell bodies and growth (reviewed in (Sofroniew et al. 2001)). Thus, our
distal axons are maintained in separate fluid environments. observation that npc–/– neurons grew normally in compart-
Thus, cell bodies/proximal axons and distal axons can be mented cultures indicated that retrograde signaling of NGF
treated and harvested separately, and axonal extension can was not severely impaired. To test whether a possible defect
be measured accurately (Campenot 1992). When viewed in NGF-dependent signal transduction in npc–/– neurons was
under the light microscope, sympathetic neurons of the masked by the high concentrations of NGF (100 ng/mL)
three npc genotypes were indistinguishable: yields of routinely used, we assessed neuronal survival in low
neurons per ganglion were comparable, npc–/– neurons concentrations of NGF, and also after deprivation of NGF
grew well in compartmented dishes and the number of for 40 h (Fig. 3). Although fewer neurons survived when
axons appeared to be the same. When distal axons were the NGF concentration was 0.5 ng/mL compared to 5 or
removed and allowed to regenerate for 6 days, no differ- 50 ng/mL, the percentage of neurons of the three npc
ences in axonal extension were observed among npc–/–, genotypes that survived at each NGF concentration was the
npc+/– and npc+/+ neurons (Fig. 2a). As confirmation that same. Thus, the sensitivity of sympathetic neuronal survival
axonal growth was the same in neurons of the 3 npc to deprivation of NGF is not dependent on NPC1.

Ó 2002 International Society for Neurochemistry, Journal of Neurochemistry, 83, 1154–1163


14714159, 2002, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1046/j.1471-4159.2002.01220.x by University Of British Columbia, Wiley Online Library on [01/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1158 B. Karten et al.

with unesterified cholesterol. Figure 4 shows that filipin


fluorescence of 12-day-old npc+/+ neurons was mainly
restricted to the plasma membrane. In contrast, a bright
punctate intracellular staining pattern, indicative of cytoplas-
mic cholesterol-laden vesicles, was evident in npc–/– neurons.
The filipin staining pattern of npc+/– neurons (Fig. 4) was
intermediate between that of npc+/+ and npc–/– neurons.
Filipin staining of neurons incubated with LDLs or high
density lipoproteins (HDLs) revealed similar fluorescence
patterns (data not shown).
Fig. 3 Dependence of survival of npc+/+, npc+/– and npc–/– neurons on In fibroblasts (Roff et al. 1992) and CHO cells (Cadigan
NGF concentration. Five-day-old neurons in 96-well plates were
et al. 1990) lacking functional NPC1, the cholesterol-laden
washed thoroughly and residual NGF was removed by incubation with
vesicles also contain the lysosomal membrane-associated
12 nM anti-NGF antibody. The neurons were then incubated for 40 h in
protein 1 (LAMP1) (Chen et al. 1985). The punctate
medium containing serum and the indicated concentrations of NGF.
The MTT survival assay was used to distinguish live and dead cells
distribution of LAMP1 was similar in neurons of all three
(stained versus not stained, respectively). Data are expressed as npc genotypes (Fig. 4). There was a pronounced colocaliza-
percentage of live cells per total cells counted and are means ± SE of tion of filipin fluorescence and LAMP1 immunoreactivity in
an experiment performed in quadruplicate. Filled bars: npc+/+, striped npc–/– neurons that was not evident in npc+/+ neurons
bars: npc+/–, open bars: npc–/–. (Fig. 4). We included an anti-murine NPC1 antibody in our
immunostaining protocol to confirm that NPC1 was
Cholesterol-laden vesicles accumulate in cell bodies expressed in npc+/+ and npc+/– neurons, but not in npc–/–
of npc–/– neurons neurons (Fig. 4). These data indicate that cholesterol accu-
We next compared the amount of unesterified cholesterol in mulates in late endosomes/lysosomes of npc–/– sympathetic
npc–/– and npc+/+ neurons by directly measuring cholesterol neurons, as has been shown in NPC1-deficient fibroblasts.
mass and by performing confocal microscopy on neurons Progesterone and the amphiphilic amine U18666A have
stained with filipin, a dye that forms a fluorescent complex been reported to induce an NPC-like phenotype (i.e.

npc +/+ npc +/- npc -/-

Filipin

LAMP1

Fig. 4 Cholesterol-filled vesicles accumu-


late in cell bodies of npc–/– neurons. Sym-
pathetic neurons from npc+/+ (left-hand
panels), npc+/– (middle panels), and npc–/–
(right-hand panels) mice were stained with
filipin and incubated with antibodies raised
NPC1 against LAMP1 or NPC1, then examined by
confocal microscopy. Scale bar ¼ 20 lm.
The experiment was performed four times
with similar results.

Ó 2002 International Society for Neurochemistry, Journal of Neurochemistry, 83, 1154–1163


14714159, 2002, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1046/j.1471-4159.2002.01220.x by University Of British Columbia, Wiley Online Library on [01/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Cholesterol accumulation in npc–/– neurons 1159

Fig. 5 Cholesterol-filled vesicles accumu-


late in cell bodies of neurons treated with
U18666A or progesterone in the presence
of LDLs. Wild-type murine neurons were
incubated for 2 days in medium containing
delipidated serum (a), or the same medium
supplemented with 20 lM progesterone and
0.1 mg/mL LDLs (b), 1.5 lM U18666A and
0.1 mg/mL LDLs (c), 20 lM progesterone
(d), or 1.5 lM U18666A (e). Filipin staining
was examined by confocal microscopy.
Scale bar ¼ 20 lm. The experiment was
performed three times with similar results.

accumulation of LDL-derived cholesterol in intracellular terol in cell bodies/proximal axons was compensated by a
vesicles) in fibroblasts (Liscum and Faust 1989; Butler et al. reduced amount of cholesterol in distal axons. We therefore
1992). To determine if these drugs also caused an accumu- measured the cholesterol content of cell bodies/proximal
lation of cholesterol in neurons we incubated wild-type
neurons for 2 days with LDLs in the absence or presence of
progesterone or U18666A. In the absence of these two drugs,
the plasma membrane stained intensely with filipin (Fig. 5a).
In contrast, incubation of the neurons with progesterone
(Fig. 5b) or U18666A (Fig. 5c) induced an intracellular
punctate filipin staining pattern that mainly colocalized with
LAMP1 (not shown) and that was similar to that seen in
npc–/– neurons (Fig. 4). Even when the neurons were
cultured in delipidated serum, U18666A or progesterone
caused accumulation of cholesterol-laden vesicles (Figs 5d
and e, respectively).

Cholesterol content of npc–/– neurons


To investigate if the increased filipin fluorescence we
observed in npc–/– neurons (Fig. 4) reflected an increase in
cholesterol content, we measured cholesterol mass in neurons
of the three npc genotypes grown in 48-well plates for
12 days. Surprisingly, the mass of cholesterol (lg choles-
terol/mg protein) in npc–/– neurons was not statistically
significantly different from that in npc+/+ neurons (Fig. 6a).
To address the possibility that the high intensity of filipin
staining was due autofluorescence of lipid-filled vesicles, we
performed confocal microscopy on npc–/– neurons in the Fig. 6 Cholesterol content and distribution in sympathetic neurons.
absence of filipin. A punctate fluorescence pattern was Sympathetic npc+/+, npc+/–, and npc–/– neurons were maintained for
12 days in 48-well plates (a) or compartmented culture dishes (b).
observed in the UV excitation range at 351 nm, similar to,
Lipids were extracted and the cholesterol content measured. In (a)
but of much lower intensity (5%) than, the filipin staining
values are given as lg cholesterol/mg protein and are means ±
in Fig. 4. Therefore, the autofluorescence exhibited by npc–/–
SE of three independent experiments with between four and six wells
neurons is only a minor contributor to the high fluorescence in each experiment. The cholesterol content of distal axons and cell
intensity observed after filipin staining. bodies/proximal axons (b) is given as percentage of the total choles-
We next considered the possibility that the reason that the terol content of cell bodies/proximal axons plus distal axons. Data
cholesterol content of npc–/– neurons as a whole was not are averages ± SE of five independent experiments. *p < 0.00075 for
increased (Fig. 6a) was because the accumulation of choles- npc–/– versus npc+/+ neurons.

Ó 2002 International Society for Neurochemistry, Journal of Neurochemistry, 83, 1154–1163


14714159, 2002, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1046/j.1471-4159.2002.01220.x by University Of British Columbia, Wiley Online Library on [01/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1160 B. Karten et al.

Fig. 7 Cholesterol-filled vesicles accumulate in 1-day-old neurons and and examined by confocal microscopy. (a) and (d) npc+/+, (b) and (e)
in neurons cultured in serum-free medium. Sympathetic neurons were npc+/–, (c) and (f) npc–/–. Scale bar ¼ 20 lm. The experiment was
plated on collagen-coated coverslips in serum-free medium. After repeated with similar results.
either 1 day (a–c) or 9 days (d–f) the neurons were stained with filipin

axons and distal axons separately in compartmented cultures.


Figure 6(b) shows that cell bodies/proximal axons of npc–/–
neurons contained significantly (p < 0.0007) more cholester-
ol than did npc+/+ and npc+/– neurons, in general agreement
with the filipin staining (Fig. 4). In contrast, the amount of
cholesterol in distal axons of npc–/– neurons was significantly
less than in npc+/+ neurons (p < 0.00075). The amount of
cholesterol/mg protein in cell bodies plus that in distal axons
of these compartmented cultures was not significantly
different in npc+/+, npc+/– and npc–/– neurons (21.3 ± 3.2, Fig. 8 The rate of incorporation of [14C]acetate into cholesterol is the
24.1 ± 4.2 and 25.5 ± 5.8 lg/mg protein, respectively), in same in npc+/+, npc+/– and npc–/– neurons. Neurons cultured in 48-well
accordance with the data (Fig. 6a) from neurons grown in plates were incubated for 24 h with [14C]acetate in medium containing
48-well plates. Moreover axonal growth and the amount of delipidated rat serum. Lipids were extracted and separated by thin-
axonal protein were the same in npc+/+ and npc–/– neurons layer chromatography. Radioactivity was measured in unesterified
cholesterol. Data are expressed as dpm/h/mg cellular protein and
(Figs 2a and b). Thus, these data show that there is not
represent means ± SE of three independent experiments.
merely an accumulation of cholesterol in cell bodies of npc–/–
neurons but that the amount of cholesterol in axons is
reduced and the distribution of cholesterol between cell npc+/– neurons (Figs 7a and b, respectively), showed a
bodies/proximal axons and distal axons is altered. pronounced punctate filipin fluorescence pattern which
persisted after 9 days in serum-free medium (Figs 7d–f).
Origin of the sequestered cholesterol Clearly, cholesterol accumulates intracellularly in cell bodies
We next attempted to determine whether the cholesterol that of npc–/– neurons in vivo during gestation and is not rapidly
accumulated in cell bodies of npc–/– neurons was derived mobilized from these vesicles. These data suggest that the
from endogenous synthesis or from exogenously supplied cholesterol accumulating in cell bodies is primarily derived
lipoproteins. Figure 4 shows that cholesterol accumulates in from endogenous synthesis.
filipin-positive vesicles in npc–/– neurons that had been
cultured in medium containing serum for 12 days. To deter- Cholesterol synthesis in sympathetic neurons
mine whether or not the formation of cholesterol-laden ves- One explanation for why cholesterol accumulates in NPC1-
icles was a consequence of the in vitro cell culture conditions deficient neurons is that the rate of cholesterol synthesis
we stained freshly isolated neurons with filipin. Even 1-day- might have been higher in npc–/– neurons than in npc+/+
old npc–/– sympathetic neurons (Fig. 7c), but not npc+/+ or neurons (Liscum and Faust 1987; Maziere et al. 1987).

Ó 2002 International Society for Neurochemistry, Journal of Neurochemistry, 83, 1154–1163


14714159, 2002, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1046/j.1471-4159.2002.01220.x by University Of British Columbia, Wiley Online Library on [01/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Cholesterol accumulation in npc–/– neurons 1161

Figure 8 shows, however, that the rate of incorporation of content of NPC1-deficient brains decreases with age (Vanier
[14C]acetate into cholesterol was similar in neurons of the 1999; Xie et al. 1999). Consequently, it was proposed that
three npc genotypes. Only background levels of radioactivity npc–/– neurons do not accumulate cholesterol (Pentchev et al.
were detected in cholesteryl esters. Some (2.5%) of the 1995). Recently, however, some elegant studies of Dietschy
labeled cholesterol was released into the medium, in amounts and coworkers have suggested that NPC1-deficient brains
that were similar in npc+/+, npc+/– and npc–/– neurons undergo a progressive loss of myelin which contains a high
(8.4 ± 1.3, 9.1 ± 1.2 and 9.4 ± 1.2 dpm/mg protein after content of cholesterol (Xie et al. 2000; German et al. 2002).
24 h, respectively). Thus, the intracellular accumulation of Xie et al. also found that the cholesterol content of brains of
cholesterol in cell bodies of npc–/– neurons cannot be one-day-old npc–/– mice was higher than of npc+/+ mice (Xie
ascribed to either a higher rate of cholesterol synthesis or a et al. 2000). Since myelin cholesterol comprises only a
decreased release of cholesterol into the medium. negligible percentage of total brain cholesterol in neonatal
mice the authors concluded that cholesterol accumulates in
NPC1-deficient neurons and/or glial cells of the central
Discussion
nervous system, as in other cell types (Xie et al. 2000).
Two important new findings concerning the function of Consequently, only in older npc–/– mice, in which extensive
NPC1 in neurons are reported in this study. First, NGF- demyelination in the brain has occurred, is there a net
dependent survival and axonal growth of NPC1-deficient decrease of brain cholesterol, compared to wild-type mice.
sympathetic neurons are normal. Second, although there is no We show that npc–/– sympathetic neurons display a bright
overall increase in the cholesterol content of npc–/– neurons, punctate pattern of filipin fluorescence, similar to that in
cholesterol accumulates in cell bodies and is partially NPC1-deficient fibroblasts and CHO cells (Sokol et al. 1988;
depleted from distal axons. Our data also suggest that the Dahl et al. 1992; Coxey et al. 1993). Filipin-positive vesi-
cholesterol accumulating in cell bodies originates primarily cles also accumulated when wild-type neurons were incuba-
from endogenous synthesis rather than from exogenously ted with LDLs in the presence of progesterone or U18666A,
supplied lipoproteins. as has been reported for fibroblasts (Butler et al. 1992;
Kobayashi et al. 1999). In contrast, direct measurement of
NGF-dependent growth of npc–/– neurons is normal cholesterol mass revealed that the cholesterol content of
The similar growth and survival characteristics of npc–/– and npc–/– and npc+/+ neurons was the same. This apparent
npc+/+ neurons indicate that NGF-dependent survival and discrepancy was resolved when we measured the mass of
growth are not impaired in npc–/– sympathetic neurons. cholesterol in cell bodies/proximal axons and distal axons
These findings are of particular interest in light of recent separately. We found that cholesterol is redistributed in npc–/–
studies in which npc–/– embryonic striatal neurons (of central neurons so that the amount of cholesterol in cell bodies of
nervous system origin) exhibited an impaired response to npc–/– neurons is greater, whereas the amount in distal axons
brain-derived neurotrophic factor (Henderson et al. 2000). In is less, than in npc+/+ neurons. These observations are
these neurons, tyrosine phosphorylation of TrkB, the receptor consistent with those of Henderson et al. (Henderson et al.
for this neurotrophin, was reduced and fewer branch points 2000) who found no difference between the cellular choles-
and shorter axons were formed in npc–/–, compared to terol content of npc–/– and npc+/+ embryonic striatal neurons.
npc+/+, neurons (Henderson et al. 2000). The different However, their study did not distinguish between cell bodies
growth characteristics and responsiveness to neurotrophins and axons and did not report whether or not filipin-positive
between striatal and sympathetic neurons from npc–/– mice vesicles accumulated in npc–/– striatal neurons.
might be because the neurotrophin receptors, TrkA and TrkB, Several studies have suggested that the cholesterol content
are activated by different growth factors, NGF and brain- of the plasma membrane of npc–/– and npc+/+ fibroblasts is
derived neurotrophic factor, respectively. Since the retro- the same (Lange et al. 2000; Garver et al. 2002; Lange et al.
grade transport of NGF after binding to the TrkA receptor is 2002) whereas other evidence has indicated that the plasma
not required for survival of sympathetic neurons (MacInnis membrane of NPC1-deficient fibroblasts (Koike et al. 1998;
and Campenot 2002), NGF-induced signal transduction Sokol et al. 1988) and CHO cells (Dahl et al. 1992) is
might still occur normally even if NPC1-deficiency affected cholesterol-poor relative to that of wild-type cells. Our
retrograde transport processes in general. observations are consistent with the idea that NPC1-defici-
ency decreases the content of cholesterol in plasma mem-
Cholesterol accumulation in NPC1-deficient neurons branes of axons since the amount of plasma membrane
One of the most obvious abnormalities in npc–/– fibroblasts relative to internal membranes is higher in axons than in
and most NPC1-deficient tissues is the accumulation of cell bodies due to the elongated shape of the neuron.
unesterified cholesterol in late endosomes/lysosomes (Sokol The observed alteration in the partitioning of cholesterol
et al. 1988; Coxey et al. 1993). In contrast, the cholesterol between cell bodies/proximal axons and distal axons of

Ó 2002 International Society for Neurochemistry, Journal of Neurochemistry, 83, 1154–1163


14714159, 2002, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1046/j.1471-4159.2002.01220.x by University Of British Columbia, Wiley Online Library on [01/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1162 B. Karten et al.

NPC1-deficient neurons leads us to speculate that the Campenot R. B. (1979) Independent control of the local environment of
anterograde movement of cholesterol from cell bodies to somas and neurites. Methods Enzymol. 28, 302–307.
Campenot R. B. (1992) Compartmented culture analysis of nerve growth,
distal axons is impaired in NPC1-deficient neurons.
in Cell–Cell Interactions: a Practical Approach (Stevenson R. B.,
Gallin W. J. and Paul D. L., eds), pp. 275–298. IRL Press, Oxford.
Origin of cholesterol accumulating in npc–/– cell bodies Carstea E. D., Morris J. A., Coleman K. G., Loftus S. K., Zhang D.,
In npc–/– fibroblasts, cholesterol-laden vesicles are more Cummings C., Gu J., Rosenfeld M. A., Pavan W. J., Krizman
evident when the cells are incubated with LDLs (Sokol et al. D. B., Nagle J., Polymeropoulos M. H., Sturley S. L., Ioannou
Y. A., Higgins M. E., Comly M., Cooney A., Brown A., Kaneski
1988). Prolonged incubation of these cells in medium
C. R., Blanchette-Mackie E. J., Dwyer N. K., Neufeld E. B., Chang
containing delipidated serum leads to a reduction in the size T.-Y., Liscum L., Strauss J. F., Ohno K., Zeigler M., Carmi R.,
and number of these vesicles suggesting that LDLs are the Sokol J., Markie D., O’Neill R. R., van Diggelen O. P., Elleder M.,
primary source of the accumulating cholesterol. In contrast, Patterson M. C., Brady R. O., Vanier M. T., Pentchev P. G. and
we found that when sympathetic neurons were cultured for Tagle D. A. (1997) Niemann–Pick C1 disease gene: homology to
mediators of cholesterol homeostasis. Science 277, 228–231.
2 weeks in lipoprotein-free medium, the intense punctate
Cham B. E. and Knowles B. R. (1976) A solvent system for delipidation
pattern of filipin fluorescence remained. Even 1-day-old of plasma or serum without protein precipitation. J. Lipid Res. 17,
npc–/– sympathetic neurons that had not been exposed to any 176–181.
exogenous cholesterol during culture were filled with Chen J. W., Murphy T. L., Willingham M. C., Pastan I. and August J. T.
cholesterol-laden vesicles, implying that these vesicles had (1985) Identification of two lysosomal membrane glycoproteins.
J. Cell Biol. 101, 85–95.
accumulated during gestation. Thus, our data indicate that the
Coxey R. A., Pentchev P. G., Campbell G. and Blanchette-Mackie E. J.
cholesterol accumulating in cell bodies of npc–/– sympathetic (1993) Differential accumulation of cholesterol in Golgi compart-
neurons is primarily derived from endogenous synthesis ments of normal and Niemann–Pick type C fibroblasts incubated
rather than from lipoproteins. This conclusion is consistent with LDL: a cytochemical freeze-fracture study. J. Lipid Res. 34,
with the idea that neurons rely heavily on cholesterol 1165–1176.
Cruz J. C. and Chang T.-Y. (2000) Fate of endogenously synthesized
synthesized endogenously. Our laboratory has previously
cholesterol in Niemann–Pick type C1 cells. J. Biol. Chem. 275,
shown that in sympathetic neurons cholesterol is synthesized 41309–41316.
in cell bodies/proximal axons, but not in distal axons (Vance Dahl N. K., Reed K. L., Daunais M. A., Faust J. R. and Liscum L. (1992)
et al. 1994; Posse De Chaves et al. 2000). An implication of Isolation and characterization of Chinese hamster ovary cells
these results is that the transport of endogenously synthesized defective in the intracellular metabolism of low density lipoprotein-
derived cholesterol. J. Biol. Chem. 267, 4889–4896.
cholesterol into distal axons of NPC1-deficient neurons
Danik M., Champagne D., Petit-Turcotte C., Beffert U. and Poirier J.
might be defective. The use of compartmented cultures of (1999) Brain lipoprotein metabolism and its relation to neuro-
neurons will permit us to test this hypothesis. As npc–/– degenerative disease. Crit. Rev. Neurobiol. 13, 357–407.
neurons survive and grow normally, at least under the culture Elleder M., Jirasek A., Smid F., Ledvinova J. and Besley G. T. N. (1985)
conditions we used, it is unlikely that NPC1 deficiency Niemann–Pick disease type C. Study on the nature of the cerebral
storage process. Acta Neuropathol. 66, 325–336.
results in a generalized defect in vesicle trafficking. Rather,
Garver W. S., Krishnan K., Gallagos J. R., Michikawa M., Francis G. A.
we suggest that the anterograde transport of the vehicles that and Heidenreich R. A. (2002) Niemann–Pick C1 protein regulates
carry cholesterol is specifically impaired. cholesterol transport to the trans-Golgi network and plasma
membrane caveolae. J. Lipid Res. 43, 579–589.
German D. C., Quintero E. M., Liang C. L., Ng B., Punia S., Xie C. and
Acknowledgements Dietschy J. M. (2001) Selective neurodegeneration, without neu-
rofibrillary tangles, in a mouse model of Niemann–Pick C disease.
This work was supported by grants from the Canadian Institutes for J. Comp. Neurol. 433, 415–425.
Health Research and the Ara Parseghian Medical Research German D. C., Liang C. L., Song T., Yazdani U., Xie C. and Dietschy
Foundation. We thank Susanne Lingrell and Russ Watts for their J. M. (2002) Neurodegeneration in the Niemann–Pick C mouse:
excellent technical assistance and Dr G. Francis (University of glial involvement. Neuroscience 109, 437–450.
Alberta) for providing human lipoproteins. Goodrum J. F. and Pentchev P. G. (1997) Cholesterol reutilization during
myelination of regenerating PNS axons is impaired in Niemann–
Pick disease type C mice. J. Neurosci. Res. 49, 389–392.
References Goodrum J. F., Brown J. C., Fowler K. A. and Bouldin T. W. (2000)
Axonal regeneration, but not myelination, is partially dependent on
Butler J. D., Blanchette-Mackie E. J., Goldin E., O’Neill R. R., Carstea local cholesterol reutilization in regenerating nerve. J. Neuro-
G., Roff C. F., Patterson M. C., Patel S., Comly M. E., Cooney A., pathol. Exp. Neurol. 59, 1002–1010.
Vanier M. T., Brady R. O. and Pentchev P. G. (1992) Progesterone Hawrot E. and Patterson P. H. (1979) Long-term culture of dissociated
blocks cholesterol translocation from lysosomes. J. Biol. Chem. sympathetic neurons. Methods Enzymol. 58, 574–584.
267, 23797–23805. Henderson L. P., Lin L., Prasad A., Paul C. A., Chang T. Y. and Maue
Cadigan K. M., Spillane D. M. and Chang T.-Y. (1990) Isolation and R. A. (2000) Embryonic striatal neurons from Niemann–Pick type
characterization of chinese hamster ovary cell mutants defective in C mice exhibit defects in cholesterol metabolism and neurotrophin
intracellular low density lipoprotein-cholesterol trafficking. J. Cell responsiveness. J. Biol. Chem. 275, 20179–20187.
Biol. 110, 295–308.

Ó 2002 International Society for Neurochemistry, Journal of Neurochemistry, 83, 1154–1163


14714159, 2002, 5, Downloaded from https://onlinelibrary.wiley.com/doi/10.1046/j.1471-4159.2002.01220.x by University Of British Columbia, Wiley Online Library on [01/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Cholesterol accumulation in npc–/– neurons 1163

Higashi Y., Murayama S., Pentchev P. G. and Suzuki K. (1993) Cere- (Scriver C. R., Beaudet A. L., Sly W. S. and Valle D., eds), pp.
bellar degeneration in the Niemann–Pick type C mouse. Acta 2625–2639. McGraw-Hill, New York.
Neuropathol. 85, 175–184. Posse De Chaves E. I., Vance D. E., Campenot R. B., Kiss R. S. and
Karten B. L. M. B., Eng H., Azumaya Y., Martin G., Lund K., Watts Vance J. E. (2000) Uptake of lipoproteins for axonal growth of
R. C., Vance J. E., Vance D. E. and Campenot R. B. (2002) sympathetic neurons. J. Biol. Chem. 275, 19883–19890.
Analytical approaches for investigating apoptosis and other Prasad A., Fischer W. A., Maue R. A. and Henderson L. P. (2000)
biochemical events in compartmented cultures of sympathetic Regional and developmental expression of the NPC1 mRNA in the
neurons, in Apoptosis Methods and Techniques, Vol. 37 (LeBlanc mouse brain. J. Neurochem. 75, 1250–1257.
A. C., ed.), pp. 163–175. Humana Press, Totowa, NJ. Roff C. F., Goldin E., Comly M. E., Blanchette-Mackie J., Cooney A.,
Kobayashi T., Beuchat M. H., Lindsay M., Frias S., Palmiter R. D., Brady R. O. and Pentchev P. G. (1992) Niemann–Pick type-C
Sakuraba H., Parton R. G. and Gruenberg J. (1999) Late endo- disease: deficient intracellular transport of exogenously derived
somal membranes rich in lysobisphosphatidic acid regulate cho- cholesterol. Am. J. Med. Genet. 42, 593–598.
lesterol transport. Nat. Cell Biol. 1, 113–118. Sattler W., Mohr D. and Stocker R. (1994) Rapid isolation of lipopro-
Koike T., Ishida G., Taniguchi M., Higaki K., Ayaki Y., Saito M., teins and assessment of their peroxidation by high-performance
Sakakihara Y., Iwamori M. and Ohno K. (1998) Decreased mem- liquid chromatography postcolumn chemiluminescence. Methods
brane fluidity and unsaturated fatty acids in Niemann–Pick disease Enzymol. 233, 469–489.
type C fibroblasts. Biochim. Biophys. Acta 1406, 327–335. Sofroniew M. V., Howe C. L. and Mobley W. C. (2001) Nerve growth
Lange Y. Ye J., Rigney M. and Steck T. (2000) Cholesterol movement in factor signaling, neuroprotection, and neural repair. Annu. Rev.
Niemann–Pick type C cells and in cells treated with amphiphiles. Neurosci. 24, 1217–1281.
J. Biol. Chem. 275, 17468–17475. Sokol J., Blanchette-Mackie E. J., Kruth H. S., Dwyer N. K., Amende
Lange Y. Ye J., Rigney M. and Steck T. L. (2002) Dynamics of lyso- L. M., Butler J. D., Robinson E., Patel S., Brady R. O., Comly
somal cholesterol in Niemann–Pick type C and normal human M. E., Vanier M. T. and Pentchev P. G. (1988) Type C Niemann–
fibroblasts. J. Lipid Res. 43, 198–204. Pick disease. Lysosomal accumulation and defective intracellular
Lange Y., Ye J. and Steck T. L. (1998) Circulation of cholesterol between mobilization of low density lipoprotein cholesterol. J. Biol. Chem.
lysosomes and the plasma membrane. J. Biol. Chem. 273, 18915– 263, 3411–3417.
18922. Taniguchi M., Shinoda Y., Ninomiya H., Vanier M. T. and Ohno K.
Liscum L. and Faust J. R. (1987) Low density lipoprotein (LDL)- (2001) Sites and temporal changes of gangliosides GM1/GM2
mediated suppression of cholesterol synthesis and LDL uptake is storage in the Niemann–Pick disease type C mouse brain. Brain
defective in Niemann–Pick type C fibroblasts. J. Biol. Chem. 262, Dev. 23, 414–421.
17002–17008. Turley S. D., Burns D. K., Rosenfeld C. R. and Dietschy J. M. (1996)
Liscum L. and Faust J. R. (1989) The intracellular transport of low Brain does not utilize low density lipoprotein-cholesterol during
density lipoprotein-derived cholesterol is inhibited in chinese fetal and neonatal development in the sheep. J. Lipid Res. 37,
hamster ovary cells cultured with 3-b-(2-(diethylamino)eth- 1953–1961.
oxy)androst-5-en-17-one. J. Biol. Chem. 264, 11796–11806. Turley S. D., Burns D. K. and Dietschy J. M. (1998) Preferential util-
Liscum L., Ruggiero R. M. and Faust J. R. (1989) The intracellular ization of newly synthesized cholesterol for brain growth in neo-
transport of low density lipoprotein-derived cholesterol is defective natal lambs. Am. J. Physiol. 274, E1099–E1105.
in Niemann–Pick type C fibroblasts. J. Cell Biol. 108, 1625–1636. Vance J. E., Pan D., Campenot R. B., Bussiere M. and Vance D. E.
Loftus S. K., Morris J. A., Carstea E. D., Gu J. Z., Cummings C., Brown (1994) Evidence that the major membrane lipids, except choles-
A., Ellison J., Ohno K., Rosenfeld M. A., Tagle D. A., Pentchev terol, are made in axons of cultured rat sympathetic neurons.
P. G. and Pavan W. J. (1997) Murine model of Niemann–Pick C J. Neurochem. 62, 329–337.
disease: mutation in a cholesterol homeostasis gene. Science 277, Vanier M. T. (1999) Lipid changes in Niemann–Pick disease type C
232–235. brain: personal experience and review of the literature. Neurochem.
MacInnis B. L. and Campenot R. B. (2002) Retrograde support of Res. 24, 481–489.
neuronal survival without retrograde transport of nerve growth Vanier M. T. and Suzuki K. (1998) Recent advances in elucidating
factor. Science 295, 1536–1539. Niemann–Pick C disease. Brain Pathol. 8, 163–174.
Maziere C., Maziere J. C., Mora L., Lageron A., Polonovski C. and Xie C., Turley S. D., Pentchev P. G. and Dietschy J. M. (1999) Cho-
Polonovski J. (1987) Alterations in cholesterol metabolism in lesterol balance and metabolism in mice with loss of function of
cultured fibroblasts from patients with Niemann–Pick disease type Niemann–Pick C protein. Am. J. Physiol. 276, E336–E344.
C. J. Inher. Metab. Dis. 10, 339–346. Xie C., Burns D. K., Turley S. D. and Dietschy J. M. (2000) Cholesterol
Naureckiene S., Sleat D. E., Lackland H., Fensom A., Vanier M. T., is sequestered in the brains of mice with Niemann–Pick type C
Wattiaux R., Jadot M. and Lobel P. (2000) Identification of HE1 as disease but turnover is increased. J. Neuropathol. Exp. Neurol. 59,
the second gene of Niemann–Pick C disease. Science 290, 2298– 1106–1117.
2301. Zervas M., Dobrenis K. and Walkley S. U. (2001) Neurons in Niemann–
Pentchev P. G., Vanier M. T., Suzuki K. and Patterson M. C. (1995) Pick disease type C accumulate gangliosides as well as unesterified
Niemann–Pick disease type C: a cellular cholesterol lipidosis, in cholesterol and undergo dendritic and axonal alterations.
The Metabolic and Molecular Basis of Inherited Disease, Vol. II. J. Neuropathol. Exp. Neurol. 60, 49–64.

Ó 2002 International Society for Neurochemistry, Journal of Neurochemistry, 83, 1154–1163

You might also like