ArticlePDF Available

Abstract and Figures

O-GlcNAcylation is an important post-translational modification of proteins and is known to regulate a number of pathways involved in cellular homeostasis. This involves dynamic and reversible modification of serine/threonine residues of different cellular proteins catalyzed by O-linked N-acetylglucosaminyltransferase (OGT) and O-linked N-acetylglucosaminidase (OGA) in antagonistic manner. We report here that decreasing O-GlcNAcylation enhances viability of neuronal cells expressing polyglutamine expanded huntingtin exon 1 protein fragment (mHtt). We further show that O-GlcNAcylation regulates the basal autophagic process and that suppression of O-GlcNAcylation significantly increases autophagic flux by enhancing the fusion of autophagosome with lysosome. This regulation considerably reduces toxic mHtt aggregates in eye imaginal discs, and partially restores rhabdomere morphology and vision in a fly model for Huntington's disease. The present study is significant in unravelling O-GlcNAcylation-dependent regulation of autophagic process in mediating mHtt toxicity. Therefore, targeting autophagic process through the suppression of O-GlcNAcylation may prove to be an important therapeutic target in Huntington disease.
O -GlcNAcylation inhibition reduces mHtt-Q97-mediated cytotoxicity ( A ) Western blot images of insoluble, aggregated form of mHtt-Q97-GFP in filter-trap assay using a slot-blot apparatus (top) or its total form resolved by immunoblotting (bottom) when expressed with either pcDNA (empty vector control), OGT or OGA, as indicated in middle. Expression of OGT and OGA was established by probing them with anti-HA and anti-Myc antibodies, respectively levels. The bar diagram above shows the fold changes in signal intensities, based on densitometric analysis, of SDS insoluble, aggregated form of mHtt-Q97- GFP (normalized to total level detected in the immunoblot; N=3; ***, p<0.001; *, p<0.1). ( B, C ) Bar diagram representing the fold change in the viability of cells expressing mHtt- Q97-GFP ( B ) or the α -synuclein mutant A40P ( C ) as measured by an MTT assay. Cells transfected with indicated constructs were processed for the measurement, and in each set the value obtained for the GFP transfected cells was considered and as 1, and the relative values obtained for indicated combinations were plotted. ( D, E ) Bar diagram showing the percentage of cells expressing mHtt-Q97-GFP ( D ) or the α -synuclein mutant A40P ( E ) with abnormal (apoptotic) nuclei (as shown in F ) as compared with cells that expressed GFP (control) when co-transfected with OGA or OGT coding constructs (in B - E , N=3; **, P<0.05; ***, P<0.005 on Student's t-test). ( F ) Representative images showing a normal (left) and an abnormal (apoptotic; right) nuclei as judged by DPAI staining (scale, 5 μM).
… 
Content may be subject to copyright.
and Subramaniam Ganesh
Parihar, Vibha Dwivedi, Subhash C. Lakhotia
Amit Kumar, Pankaj Kumar Singh, Rashmi
exon1 protein fragment
from cytotoxicity mediated by huntingtin
Decreased O-linked GlcNAcylation protects
Molecular Bases of Disease:
published online March 19, 2014J. Biol. Chem.
10.1074/jbc.M114.553321Access the most updated version of this article at doi:
.JBC Affinity SitesFind articles, minireviews, Reflections and Classics on similar topics on the
Alerts:
When a correction for this article is posted When this article is cited
to choose from all of JBC's e-mail alertsClick here
http://www.jbc.org/content/early/2014/03/19/jbc.M114.553321.full.html#ref-list-1
This article cites 0 references, 0 of which can be accessed free at
by guest on March 20, 2014http://www.jbc.org/Downloaded from by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
1
Decreased O-linked GlcNAcylation Protects from Cytotoxicity Mediated by Huntingtin
Exon1 Protein Fragment*
Amit Kumar1,3,4, Pankaj Kumar Singh1,3,5 Rashmi Parihar1,3, Vibha Dwivedi2,3, Subhash C.
Lakhotia2 and Subramaniam Ganesh1,6
From the 1Department of Biological Sciences and Bioengineering, Indian Institute of
Technology, Kanpur, India;
2Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi,
India
3These authors contributed equally and should be considered as joint first authors
Running Title: Decreasing O-linked GlcNAcylation increases basal autophagic flux
6To whom correspondence should be addressed: Subramaniam Ganesh, Department of
Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016,
India; Tel (+91) 512 259 4040; Fax (+91) 512 259 4010; Email: sganesh@iitk.ac.in
Key words: O-GlcNAcylation; Autophagy; Huntington’s disease
Background: Earlier reports indicate that
O-GlcNAcylation might be protective in
neurodegenerative disorders.
Results: Suppressing O-GlcNAcylation
modulates autophagy to enhance the
viability of neuronal cells expressing
cytotoxic mutant huntingtin exon 1 protein
(mHtt).
Conclusion: O-GlcNAcylation regulates
the clearance of mHtt by modulating the
fusion of autophagosomes with lysosomes.
Significance: This regulatory mechanism
emerges as a novel therapeutic strategy for
Huntington’s disease.
ABSTRACT
O-GlcNAcylation is an important
post-translational modification of
proteins and is known to regulate a
number of pathways involved in cellular
homeostasis. This involves dynamic and
reversible modification of
serine/threonine residues of different
cellular proteins catalyzed by O-linked
N-acetylglucosaminyltransferase (OGT)
and O-linked N-acetylglucosaminidase
(OGA) in antagonistic manner. We
report here that decreasing O-
GlcNAcylation enhances viability of
neuronal cells expressing polyglutamine
expanded huntingtin exon 1 protein
fragment (mHtt). We further show that
O-GlcNAcylation regulates the basal
autophagic process and that suppression
of O-GlcNAcylation significantly
increases autophagic flux by enhancing
the fusion of autophagosome with
lysosome. This regulation considerably
reduces toxic mHtt aggregates in eye
imaginal discs, and partially restores
rhabdomere morphology and vision in a
fly model for Huntington’s disease. The
present study is significant in
unravelling O-GlcNAcylation-dependent
regulation of autophagic process in
mediating mHtt toxicity. Therefore,
targeting autophagic process through
the suppression of O-GlcNAcylation
may prove to be an important
therapeutic target in Huntington
disease.
INTRODUCTION
O-GlcNAcylation is a glucose
dependent post-translational modification
(PTM). When glucose enters the cell,
approximately 5% of it enters into the
http://www.jbc.org/cgi/doi/10.1074/jbc.M114.553321The latest version is at
JBC Papers in Press. Published on March 19, 2014 as Manuscript M114.553321
Copyright 2014 by The American Society for Biochemistry and Molecular Biology, Inc.
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
2
Hexosamine biosynthetic pathway (HBP)
through a series of metabolic
transformation and finally gets
transformed into uridine 5’-diphospho-N-
acetyl glucosamine (UDP-GlcNAc) (1).
This final product of HBP, amongst other
functions, acts as a substrate of O-linked
GlcNAcylation and is utilized by two
antagonistic enzymes, O-linked N-
acetylglucosaminyltransferase (OGT) and
O-linked N-acetylglucosaminidase (OGA),
which regulate the dynamic modification
of different nucleo-cytoplasmic proteins.
OGT catalyzes the addition of a single
GlcNAc moiety to serine and/or threonine
sites of various proteins while OGA
removes the same (2). O-GlcNAcylation
has been shown to regulate many vital
biological processes such as replication,
transcription, translation, stress response,
nutrient response, unfolded protein
response, and intra-cellular protein
trafficking (3-5). It is also emerging as an
important regulatory mechanism in a
number of complex diseases such as
diabetes, cancer, cardiovascular diseases,
and in ageing (5). OGT activity has been
reported to be about 10 times more
enriched in brain as compared to other
tissues like liver, muscle, adipose and heart
(6) and has been shown to glycosylate
many proteins linked to neurodegenerative
diseases such as, amyloid precursor
protein, β amyloid associated protein (7),
microtubule associated tau protein (8),
synapsin (9) and neurofilament proteins
(10). Recently, OGT was reported to play
a protective role in Alzheimer’s disease
(11) and is speculated to be protective in
other neurodegenerative disorders such as
Huntington’s disease, Parkinson’s disease
and Amyotrophic Lateral Sclerosis (ALS).
Huntington’s disease is a
neurodegenerative disorder characterized
by the formation of intracellular
aggregates of mutant huntingtin (mHtt)
(12, 13). The normal huntingtin gene codes
for the huntingtin protein, which usually
has up to 34 glutamine coding (CAG)
repeat (12, 13). The huntingtin protein
having up to 34 glutamine repeats
represents a normal functional protein,
while expansion of CAG repeats coding
for >40 glutamine as a repeat track results
in a dominant mutation, as a consequence
of which the mutant Htt loses its proper
folding state, tends to aggregate and
becomes cytotoxic (12, 13). The wild-type
huntingtin protein plays important roles in
normal functioning of brain such as
vesicular transport, neuronal gene
transcription, BDNF production (14), and
may also function as an anti-apoptotic
protein (15). The mHtt aggregates interfere
with normal synaptic transmission (16),
impair axonal transport of mitochondria
(17), sequester crucial transcription factors
(18) and hamper their functioning.
O-GlcNAcylation is a nutrient
sensitive protein modification. With the
emerging understanding of the important
roles of O-GlcNAcylation in various
neurodegenerative disorders along with
reports about glucose-dependent regulation
of protein clearance machineries (19, 20)
and that of protein aggregation mediated
toxicity (21, 22), we aimed to explore the
role of this glucose-dependent post-
translational modification in the regulation
of mHtt-mediated toxicity and its
clearance. We report here that suppression
of O-GlcNAcylation increases basal
autophagy flux by enhancing
autophagosome-lysosome fusion and helps
in the clearance of toxic aggregates of
mutant huntingtin exon 1 coded protein,
thereby increasing survival and
suppressing the degenerative phenotypes
in cellular and Huntington fly models,
respectively.
EXPERIMENTAL PROCEDURES
Reagents and antibodies
Cell culture
media and drugs (Azaserine, Glucosamine,
3-Methyladenine, and Bafilomycin A1)
were purchased from Sigma Aldrich Pvt.
Ltd., India. Polyfect transfection reagent
was from QIAGEN India Pvt. Ltd., India.
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
3
Antibodies were procured from following
sources: Anti-LC3 and Anti-Myc for
immunostaining (Cell Signaling
Technology, USA); Anti-p62 (Enzo Life
Sciences, USA); Anti-GFP and Anti-Myc
for immunoblots (Roche, India); Anti-HA
(used in experiments done in Neuro2A
cell) (Sigma-Aldrich Pvt. Limited, India),
Anti-O-GlcNAc and Anti-γ-tubulin
(Sigma-Aldrich Pvt. Limited, India).
Rabbit polyclonal anti-haemagglutinin
(used in experiments with Drosophila)
(Santa Cruz, USA), Secondary antibodies
were procured from Jackson
ImmunoResearch Inc., USA, except Anti-
rabbit conjugated with Cy3 (Sigma-
Aldrich, India) and Alex-Fluor 488
(Molecular Probes, USA).
Expression constructs
Mammalian
expression constructs were obtained from
following sources: The GFP-tagged
truncated Huntingtin Q97 expression
vector was generously provided by Dr
Lawrence Marsh (University of California,
Irvine, CA, USA). Plasmid coding for
OGA (Myc-tagged) was a kind gift from
Dr John A Hannover (NIDDK, National
Institutes of Health, USA), the HA-tagged-
OGT was gifted by Dr Gerald W. Hart
(Johns Hopkins University School of
Medicine, Baltimore, USA), the mRFP-
GFP-LC3 construct was gifted by Dr T.
Yoshimori (National Institute for Basic
Biology, Okazaki, Japan), and construct
coding for the mutant form of α-synuclein
as GFP fusion was a gift from Dr. Peter
Lansbury (Harvard Medical School, USA).
Cell culture, treatment and transfection
The experiments were conducted in the
murine neuroblastoma cell line Neuro2A
under normal glucose conditions (25 mM).
Neuro2A cells were grown in Dulbecco’s
modified Eagle’s medium (Sigma-Aldrich
Pvt. Limited, India) supplemented with
10% (v/v) fetal calf serum, 100 U/ml
penicillin and 100 µg/ml streptomycin.
The cells were treated with 40 µM
azaserine (inhibitor of O-GlcNAcylation),
10 mM glucosamine (inducer of O-
GlcNAcylation), 100 nM Bafilomycin A1
(inhibitor of the fusion of autophagosomes
with lysosomes) and 10 mM 3-
Methyladenine (3MA; an inhibitor of
autophagosome formation). The cells were
transiently transfected with expression
constructs at around 50% of confluence
using PolyFect Transfection Reagent
(QIAGEN India Pvt. Ltd) as recommended
by the manufacturer. Under these
conditions the transfection efficiency was
consistent and around 70% as assessed by
microscopic observation of the
fluorescence positive cells transiently
expressing the GFP-tagged protein. In all
the experiments, the cells were harvested
at 36 hrs post-transfection and wherever
required, treatment with the
pharmacological agents was given for the
last 12 hrs unless stated otherwise.
Fly stocks and rearing condition All fly
stocks were maintained under un-crowded
condition at 240C±10C. For each
experiment, regular or azaserine
(250µg/ml) supplemented food was
prepared from the same batch. Using the
w1118; UAS-httex1p Q93/CyO (20) and
w1118; GMR-GAL4 (21) fly stocks,
appropriate genetic crosses were set to
obtain w1118; UAS-httex1p Q93/GMR-
GAL4 (GMR-GAL4>UAS-httex1p)
progeny. The GMR-GAL4 driver targets
expression of the UAS-httex1pQ93
transgene in developing eye discs (23) and
thereby induce retinal neurodegeneration
(24). In some cases, Oregon R+ stock was
used as wild type. Freshly hatched larvae
for a given experiment were derived from
a common pool of eggs of the desired
genotype and reared in parallel on regular
or azaserine supplemented food.
We also reared larvae on food
supplemented with glucosamine (1 mg/ml,
10 mg/ml or 25 mg/ml). However, in each
case, all the larvae died before reaching 3rd
instar stage and therefore, no further
studies on the effect of glucosamine on
polyglutamine (polyQ) degeneration in the
fly model could be carried out.
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
4
Immunostaining
Cells on coverslip were
fixed with 4% paraformaldehyde in 1X
PBS for 20 minutes followed by
permeabilization for 5 minutes in 1X PBS
with 0.05% Triton X-100. The expression
of Myc-OGA and HA-OGT was checked
by probing with Anti-Myc or Anti-HA
antibodies followed by FITC or TRITC
conjugated secondary antibodies,
respectively. Nuclei were counterstained
with 10µM 4’,6-diamidino-2-phenylindole
(DAPI). Images were obtained with a
Nikon (Japan) Eclipse 80i fluorescence
microscope using a 10X or 40X objective
lens.
Eye discs from wandering late third
instar GMR-GAL4>UAS-httex1p Q93
larvae reared on normal or azaserine
supplemented food were dissected and
immunostained as described previously
(25) with the anti-HA (1:80 dilution,
Santa-Cruze). Chromatin was
counterstained with DAPI. Immuno-
fluorescence stained eye discs were
examined with a Zeiss LSM 510 Meta
confocal microscope using appropriate
lasers, dichroics and filters.
Cell death assay For the MTT assay
cells were treated with azaserine or
glucosamine for 12 hrs or transfected for
36 hrs and thereafter, cells were incubated
with 0.5 mg/ml MTT (thiazolyl blue
tetrazolium bromide) (Sigma-Aldrich Pvt.
Limited, India) and chased for 2 hrs. After
removal of the medium, cells were
incubated with DMSO (100%) for 10 min
to dissolve formazon crystals. The change
in optical density was recorded through
spectrophotometer at λ570nm against
background reading at λ650nm.
Alternatively, treated or transfected cells
were fixed, permeabilized and stained with
DAPI as mentioned for immunostaining,
and the apoptotic nuclei were scored in a
blinded fashion as reported earlier (26).
Quantification of LC3-positive cytoplasmic
puncta - Cells transiently expressing the
tandem mRFP-GFP-LC3 construct were
fixed, and the fluorescence images of
about 50 cells for each set were examined
using a Zeiss AxioImager 2 microscope
outfitted with an ApoTome accessory. The
green, red and yellow puncta in the
captured images were quantified using the
Colocalization Macro in ImageJ software,
as described (27)
Immunoblotting
Protein samples were
resolved on 6-12% SDS-PAGE as required
and transferred to nitrocellulose membrane
(MDI, India). Thereafter, the membranes
were blocked with either 5% non-fat dry
milk powder or 5% BSA in 1X TBST and
probed sequentially with the desired
primary and secondary antibodies at their
recommended dilutions followed by
detection with a chemiluminescent
detection kit (Supersignal West PICO,
Pierce, USA).
Filter-trap assay The filter-trap assay
was carried out essentially as described by
Juenemann et al. (28). Briefly, the pellet
fraction of the cell lysate was suspended in
the benzonase buffer (1 mM MgCl2, 50
mM Tris/HCl pH 8.0), and treated with a
RNAse/DNAse cocktail (50 U each;
Fermentas) and incubated for 1 hr at 37oC.
The reaction was arrested with the addition
of 2x termination buffer (40 mM EDTA, 4
% SDS, 100 mM DTT), and 50 μg of the
sample was mixed in 2% SDS buffer (2 %
SDS, 150 mM NaCl, 10 mM Tris/HCl pH
8.0) and filtered through a 0.2 μm pore
size cellulose acetate membrane (GE
Healthcare Life Sciences, USA) using a
slot blot apparatus (Bio-Rad Indian Pvt.
Ltd., India). The filter membrane was used
for immunodetection as described for the
immunoblot.
Proteasome activity assays - Cells, that
were either transfected or treated with
indicated drugs (12 h), were harvested in
lysis buffer (1x PBS, 0.1% Triton X100,
0.5% NP40) and the cleared lysate was
used for the proteasome activity assay
using a fluorogenic proteasome substrate
(Suc-Leu-Leu-Val-Tyr-AMC;
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
5
Calbiochem). Briefly, 10 µg of protein for
each sample was incubated in a reaction
buffer and the generation of fluorescent
signal was measured using a
spectrofluorometer (Parkin Elmer) as
recommended by the manufacturer.
Reactions in the presence of the
proteasomal blocker, MG132, served as
control.
Pseudopupil Analysis Heads of 1 day old
GMR-GAL4>UAS-httex1p Q93 flies,
reared since the 1st instar larval stage on
normal or azaserine supplemented food
were decapitated and the arrangement of
photoreceptor rhabdomeres in the
ommatidia of compound eyes was
visualized by the pseudopupil technique
(29) using 63X (NA= 1.4) oil objective on
a Nikon E800 microscope and the images
were recorded with a Nikon DXM 1200
digital camera. The total number of flies
observed for each group was 50.
Phototaxis Assay Phototaxis of adult
flies was assayed using a Y maze
consisting of a Y shaped glass tube of 12
mm internal diameter and 30 cm length of
each arm. Twenty replicates, each with 10
flies, were carried out for each feeding
regime and age of flies. Wild type Oregon
R+ flies were used as positive control. The
same sets of flies were used for phototaxis
assay on days 0, 5, 10 and 15.
Statistical analysis Sigma Plot 11.0
software was used for statistical analysis.
For cell biology assays, data were
analyzed by two-tailed, unpaired Student’s
t-test. For assays involving flies, one-way
ANOVA was performed for comparison
between the control and formulation-fed
samples. Pooled data are expressed as
mean ± S.E. of means of the different
replicates of the experiment.
RESULTS
Global suppression of O-linked
glycosylation reduces the aggregation
propensity and cytotoxicity of mutant
Huntingtin in a cellular model
Based on
the previous findings (21, 22), we were
interested in exploring the role of O-
GlcNAcylation in suppressing the
cytotoxicity caused by aggregate-prone
proteins. For this, we used a mammalian
expression construct that codes for the
OGT or OGA, the two proteins which
work antagonistically to regulate the O-
linked protein glycosylation. Transient
expression of OGT in the murine
neurobalstoma cell line Neuro2A resulted
in increased global O-GlcNAcylation
while overexpression of OGA led to a
reduction in global O-GlcNAcylation (Fig.
1A). To check if O-GlcNAcylation could
alter the aggregate forming propensity of
mutant huntingtin, we co-expressed OGA
or OGT with an expression construct
coding for the amino terminal huntingtin
protein having 97 polyglutamine repeat
tagged with green fluorescence protein
(mHtt-Q97-GFP) in Neuro2A. Cells that
expressed only the mHtt-Q97-GFP served
as control (co-transfected with the empty
vector pcDNA). As shown in Fig. 1B and
1D, co-expression of OGA resulted in a
significant reduction in the number of
transfected cells showing the mHtt-Q97-
GFP-positive aggregates as compared to
the control cells that were co-transfected
with the empty vector, pcDNA. On the
other hand, OGT co-expression resulted in
a higher proportion of cells with the mHtt-
Q97-GFP aggregates (Fig. 1B and 1D).
Co-expression of OGA or OGT did not
affect the transfection efficiency of the
mHtt-Q97-GFP coding construct (see the
inlet Fig. 1D). Similarly, there was no
significant difference in cell survival when
OGA or OGT was expressed alone as
compared to cells that were transfected
with an empty vector (Fig. 1C). To test
further whether the reduction in the global
O-GlcNAcylation helps the cell to reduce
the aggregation of mHtt-Q97-GFP, we
evaluated the total and SDS-insoluble
forms of mHtt-Q97-GFP by immunoblot
and the filter-trap assay, respectively (Fig.
2A). Consistent with our observations on
mHtt aggregates in situ, co-expression of
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
6
OGA led to a significant reduction in the
level of SDS-insoluble form of mHtt-Q97-
GFP when compared with that in cells co-
expressing OGT or only the mHtt-Q97-
GFP (pcDNA control; Fig. 2A). Co-
expression of OGA or OGT did not show
significant change in the total level of
mHtt-Q97-GFP in the immunoblots (Fig.
2A). We also found that co-expression of
OGA, but not of OGT, resulted in a
significant reduction in mHtt-Q97-GFP-
mediated cell death, as measured by MTT
assay (Fig. 2B), and also by scoring
apoptotic nuclei (Fig. 2D). To check
whether the protective effect of OGA was
limited to mtHtt-Q97-GFP, or whether
OGA can ameliorate the toxicity of other
disease associated cytotoxic proteins, we
expressed Parkinson’s disease associated
α-synuclein mutant A30P protein (26)
either alone or along with OGA or OGT
and measured the cell viability by MTT
assays as well as by counting the apoptotic
nuclei. As shown in Fig. 2C and 2E, OGA,
but not OGT, was able to confer protection
against the toxicity of the A30P mutant,
suggesting that the OGA-mediated
protective response could be a generic
effect of O-GlcNAcylation, and is not
specific to mtHtt-Q97-GFP. Taken
together, our results suggest a causal role
for O-GlcNAcylation in modulating the
level of insoluble, aggregated mutant
huntingtin and its cytotoxicity.
Inhibition of O-GlcNAcylation enhances
autophagy Our next aim was to identify
the mechanism by which suppression of
O-GlcNAcylation reduces the level of
cytotoxic insoluble form of mHtt-Q97-
GFP. Since autophagic process is known
to clear the aggregated proteins (30, 31),
we were interested in testing the impact of
O-GlcNAcylation in basal autophagic
process. For this, Neuro2a cells were
transfected with the expression construct
coding for OGT or OGA or with an empty
vector (pcDNA control). At 36 hrs post-
transfection, the cells were harvested and
levels of two autophagic marker proteins,
LC3 and p62, were evaluated. As shown in
Fig. 3A, transient overexpression of OGA
led to a reduction in the level of both p62
and LC3II, suggesting that suppression of
O-GlcNAcylation resulted in an enhanced
autophagic flux. To further confirm that
the observed effect is indeed because of
the changes in global O-GlcNAcylation,
we examined the autophagic process after
treating the cells with, azaserine, which
inhibits glutamine fructose-6-phosphate
amidotransferase, one of the key enzymes
of hexosamine biosynthesis pathway and
thereby inhibits O-GlcNAcylation (32,
33). As shown in Fig. 3B, treatment of
Neuro2A cells with azaserine for 12 hrs
resulted in a significant reduction in global
O-GlcNAcylation levels. As was observed
for OGA expression, azaserine also led to
a reduction in the level of the autophagic
markers LC3II and p62 (Fig. 3C),
confirming that a reduction in the cellular
O-GlcNAcylation level correlates with
increased levels of basal autophagic flux.
To further confirm that the observed effect
of azaserine on autophagic process is
indeed through O-GlcNAcylation process,
we treated cells both with azaserine and
glucosamine and looked at the level of
LC3II and p62. Glucosamine is known to
rescue the effect of azaserine on O-
GlcNAcylation process hence the double
treatment should rescue the effect of
azaserine on the autophagic process (Fig.
3B). As shown in Fig. 3C, azaserine-
glucosamine treatment increased the level
of LC3II and p62 as compared to only
azaserine treatment, confirming that the
level of autophagic induction inversely
correlate with O-GlcNAcylation level.
Our next aim was to identify the key
step through which the O-GlcNAcylation
regulates the autophagic process. The
reduction in the level of the autophagy
marker LC3II upon depletion of
glycosylation may be because either (i) the
autophagosome formation is inhibited
(inhibition of autophagy initiation), or (ii)
enhanced degradation of LC3II (increased
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
7
autophagic flux) via lysosome since LC3
itself is an autophagy substrate (34). Our
observation that the cellular level of
another autophagic substrate, p62, was
also at lower levels upon OGA
overexpression suggests that the second
possibility is more likely. Therefore, we
checked whether inhibition of fusion of
autophagosome with the lysosome would
rescue the level of LC3II and p62. For this,
the cells were transfected with expression
construct coding for OGA of OGT and
then were treated with bafilomycin A1
(BafA1), an inhibitor of autophagosome
lysosome fusion (34) for 12 hrs and the
cellular level of LC3II and p62 was
evaluated. As shown in Fig. 4A, we found
that the BafA1-mediated inhibition of
autophagosome-lysosome fusion led to an
increase in the level of both LC3II and p62
even in those cells that overexpressed
OGA or OGT. Very similar observations
were made when the glycosylation was
inhibited by azaserine treatment (Fig. 4B).
To further confirm that suppression of O-
GlcNAcylation indeed increases the
autophagy flux, we utilized the tandem
mRFP-GFP-LC3 expression construct
whose expression product is known to
show difference in pH sensitivity and has
been widely used to monitor the
autophagic process (35). For this the
Neuro2A cells were transiently transfected
with the mRFP-GFP-LC3 tandem
construct and empty vector (pcDNA), or
along with the expression vector coding
for OGA or OGT, and scored the
colocalization of green and red signals in
the cytoplasmic LC3-positive puncta, and
also the number of green and red puncta.
Here, autophagosomes are visible as
yellow puncta and autophagolysosomes
(post-lysosomal fusion) as red puncta (35).
As shown in Fig. 5, co-expression of OGA
led to a significant increase in the fraction
of red/green-positive LC3 puncta while no
such difference was noted for OGT.
Similarly, there was a significant increase
in the LC3 puncta that were positive only
for red fluorescence (Fig. 5), suggesting
that suppression of O-GlcNAcylation did
enhance the autophagy flux.
Next, we tested whether the
reduction in the level of insoluble fraction
of mutant huntingtin seen in O-
GlcNAcylation deprived-condition is due
to an enhanced autophagy flux. As shown
in Fig. 6A, BafA1 treatment led to an
increase in the level of insoluble fraction
of the mutant huntingtin even in the OGA
overexpressing cells, suggesting that
decreased O-GlcNAcylation promotes the
clearance of the aggregate-prone protein
by enhancing autophagosome-lysosome
fusion. Finally, to demonstrate that
autophagy is the mechanism through
which O-GlcNAcase is able to protect cells
from huntingtin aggregates, we treated
cells that coexpress mtHtt-Q97-GFP and
OGA with an autophagy inhibitor, 3-
Methyladenine (3-MA). As shown in Fig.
6B, 3-MA treatment led to a significant
increase in the insoluble form of mtHtt-
Q97-GFP even when OGA was co-
expressed, suggesting that the protective
effect conferred by OGA is indeed through
the autophagic process.
Having shown an indirect correlation
between O-GlcNAcylation and autophagic
flux, we checked possible effect of O-
GlcNAcylation on proteasomal activity.
For this cells that transiently expressed
OGA or OGT or that were treated with
azaserine or glucosamine were assayed for
proteasomal activity. As shown in Fig. 7A,
transient overexpression of OGT or OGA
led to a significant reduction in the
proteasomal activity. Treatment of cells
with azaserine or glucosamine did not
significantly alter the activity (Fig. 7B),
suggesting that the O-GlcNAcylation-
dependent clearance of mutant huntingtin
observed in our model could be primarily
through the autophagic process.
Azaserine feeding reduces mutant
huntingtin aggregation in the larval eye
discs of Drosophila Having found that
azaserine treatment reduces the aggregates
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
8
of mutant huntingtin in the mammalian
cell line, we next tested whether similar
effect could also be seen in vivo, for which
we used the fly model of Huntington’s
disease (23). We reared wild type and
GMR-GAL4>httex1p Q93 larvae from the
1st instar stage onwards on food
supplemented with azaserine (250 µg/ml).
It is known (36, 37) that GMR-GAL4
driven expression of the mutant huntingtin
protein leads to accumulation of polyQ
inclusion bodies posterior to the
morphogenetic furrow in late 3rd instar
larval eye discs (Fig. 8A, B). We found
that azaserine feeding substantially
reduced the accumulation of mHtt protein
so that in ~57% of the eye discs (n = 30)
from azaserine-fed larvae, the aggregates
were nearly absent behind the
morphogenetic furrow (Fig. 8C, D) while
in the remaining discs immunostaining
was less than that in the eye discs (n = 29)
from larvae reared on regular diet (Fig.
6A, B). Western blotting for detection of
polyQ protein levels in heads of day 1 old
GMR-GAL4>httex1p Q93 flies further
confirmed that azaserine feeding reduced
the level of polyQ protein (Fig. 8E, F).
Azaserine feeding partially restores the
rhabdomere morphology and suppresses
the progressive loss of vision in GMR-
GAL4>UAS-httex1p Q93 expressing flies
The external eye morphology and vision
of freshly eclosed GMR-GAL4>UAS-
httex1p Q93 flies is near normal. However,
these flies show a progressive age-
dependent degeneration, becoming almost
completely blind by 10 days (36-38). As
known from earlier studies (36-38), the
eye surface of GMR-GAL4>UAS-httex1p
Q93 flies did not show any appreciable
change with age in any of the feeding
regimes (not shown). However, as also
reported earlier (36, 38), the pseudopupil
images of rhabdomeres of 1 day old GMR-
GAL4>UAS-httex1p Q93 expressing flies
fed on normal diet showed severely
degenerated rhabdomeres so that unlike
the stereotyped pattern of rhabdomeres in
pseudopupil image of eyes of wild type
flies (Fig. 9A), no distinct rhabdomeres
were seen in their eyes (Fig. 8B).
Interestingly, GMR-GAL4>UAS-httex1p
Q93 flies reared on the azaserine
supplemented food displayed at least some
organized rhabdomere-like structures in
~60% flies (Fig. 8C).
Expression of UAS httex1p Q93 in
eye cells with GMR-GAL4 driver causes
progressive neuronal degeneration of the
photoreceptor neurons so that the flies lose
their vision as they age (36, 38). To
examine whether the azaserine mediated
restoration of rhabdomeric organization
improved the vision of flies, we tested the
functionality of vision in 5, 10 and 15 days
old flies (wild type and GMR-
GAL4>httex1p Q93) by the phototaxis
behavioral assay, which examines the
choice of flies to move between
illuminated and dark chambers. While
nearly all wild-type flies of different ages
moved to the illuminated chamber
(positive phototaxis), the GMR-
GAL4>httex1p Q93 flies reared on normal
food progressively lost their vision such
that the proportion of flies selecting the
lighted chamber declined with age (Fig.
9D). By day 10, these flies became nearly
blind since they moved randomly between
the dark and light chambers (Fig. 9D).
Significantly, a greater proportion of
GAL4>httex1p Q93 flies reared on
azaserine-supplemented food continued to
move to the illuminated chamber even on
day 15 (Fig. 9D). Thus azaserine feeding
partially restored the vision in
GAL4>httex1p Q93 flies so that the
proportion of flies selecting the
illuminated chamber was significantly
higher on each day of phototaxis assay
than in those grown on normal food (Fig.
9D).
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
9
DISCUSSION
Dynamic modification of Ser/Thr residues
of proteins by O-linked N-
acetylglucosamine (O-GlcNAc) is an
important post-translational modification
for cellular signaling (1, 3, 39). More than
five hundred proteins, involved in diverse
cellular functions including the
transcription, translation, metabolism and
stress response, have been identified to
undergo this modification (1, 3, 39). It is
significant that about 270 of these proteins
are known in the brain tissue alone (40).
Therefore it is not surprising that aberrant
O-GlcNAcylation is associated with
various disorders, including the
neurodegenerative disorders (3-11).
A common pathological feature of
many neurodegenerative diseases
including Alzheimer’s, Parkinson’s and
Huntington’s diseases, is the
accumulation/aggregation of one or more
proteins in different regions of brain,
which is believed to underlie
neurodegeneration (12, 37, 41). These
proteotoxic aggregates are cleared by
coordinated action of cellular proteolysis
system (UPS and autophagy-lysosomal
pathways) and molecular chaperones (12,
30). Although the regulation of UPS by O-
GlcNAcylation is fairly understood, there
are contrasting reports about a protective
role of O-GlcNAcylation in
neurodegeneration. For example, it is
shown that while O-GlcNAcylation of
ubiquitin-activating enzyme E1 promotes
ubiquitination (42) and is thus expected to
enhance protein degradation, the same
modification in Rpt2 ATPase subunit of
the proteasome inhibits its ATPase activity
and suppresses proteasome function (43),
which would lead to the accumulation of
ubiquitinated proteins. Interestingly, it is
shown that elevated O-GlcNAcylation in
brain inhibits proteasome function and
promotes neuronal apoptosis (44). We find
that overexpression of either OGT or OGA
led to significant reduction in the
proteasome activity in our cellular model
and this corroborates well with a recent
report on proteasomal function in C.
elegans mutants for OGT or OGA (45).
However, we did not find any difference in
the proteasome activity when the cells
were treated with azaserine or glucosamine
for the duration and concentration used,
suggesting that the level and/or activity of
OGT and OGA, rather than flux through
the HBP alone, are more critical in
modulating the activity of proteasome. In
view of these observations, and existing
reports that accumulation of protein
aggregates blocks proteasome function
(46, 47), it appears that proteasome alone
might not be sufficient to clear these
aggregates. This notion is strengthened
with the emerging understanding of the
role of autophagy in degradation of such
aggregates in cell and animal models (12,
27, 48), identification of novel regulators
of autophagy that help in the clearance of
toxic protein aggregates is important.
Considering the established fact that O-
GlcNAcylation acts as nutrient sensor (3,
49), and the role of nutrients (serum amino
acid, glucose) in regulation of the
autophagic process (50, 51), we
hypothesized that changes in O-
GlcNAcylation level might modulate
autophagic process. Interestingly, we
found here that inhibition of O-
GlcNAcylation, either by overexpressing
OGA or by azaserine treatment, decreased
the polyQ aggregation by promoting their
clearance via autophagy.
In agreement with the results of our
in vitro cell culture model, our studies on
the in vivo fly model also revealed that
azaserine feeding resulted in the
improvement in Drosophila eyes
expressing mutant huntingtin at cellular,
phenotypic, as well as at functional level in
the form of reduced aggregation of mHtt,
improved rhabdomere organization and
improved vision, respectively. Since
glucosamine was highly toxic to larvae
even at a very low concentration, we could
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
10
not examine the effects of elevated levels
of O-GlcNAcylation on polyQ toxicity in
the fly model. The decrease in
proteotoxicity on azaserine feeding might
involve either the inhibition of toxic
proteins synthesis or its enhanced
clearance through the degradation
machinery of cell. Several recent findings,
including our present in vitro findings,
suggest a greater role of enhanced
clearance of toxic proteins by azaserine.
Thus the reduced polyQ aggregate load
seen in azaserine fed GAL4>httex1p Q93
larval eye discs and adult heads is likely to
be due to their enhanced clearance, which
in turn results in partial restoration of eye
structure and function.
Azaserine has already been reported
to decrease the level of amyloid deposition
in pancreatic islets of mouse model of
diabetes (52) which indicates the
possibility of improvement in protein
clearance machinery and thereby reducing
the accumulation of amyloid deposits. Our
observations that inhibition O-
GlcNAcylation induces the clearance of
protein aggregates by enhancing the
autophagic process is in agreement with a
recent finding that cardiac O-
GlcNAcylation regulates autophagic
signaling in rat model of type II diabetes
(53). Interestingly, a recent report, which
appeared while this manuscript was in
preparation, by Wang et al. (54) in C.
elegans model of human
neurodegenerative diseases also indicates
that the suppression of O-GlcNAcylation
decreases neurodegeneration. Taken
together, our in vitro and in vivo findings
indicate that inhibition of O-
GlcNAcylation stimulates autophagy and
thereby reduces the load of proteotoxic
huntingtin aggregates and provides
protection from neurodegeneration.
REFERENCES
1. Love, D.C., and Hanover, J.A. (2005) The hexosamine signaling pathway: deciphering
the "O-GlcNAc code". Sci STKE. 2005, re13.
2. Vocadlo, D.J. (2012) O-GlcNAc processing enzymes: catalytic mechanisms, substrate
specificity, and enzyme regulation. Curr. Opin. Chem. Biol. 16, 488-497.
3. Butkinaree, C., Park, K., and Hart, G. W. (2010) O-linked beta-N-acetylglucosamine (O-
GlcNAc): Extensive crosstalk with phosphorylation to regulate signaling and
transcription in response to nutrients and stress. Biochim. Biophys. Acta. 1800, 96-106.
4. Chatham, J. C., and Marchase, R. B. (2010) Protein O-GlcNAcylation: A critical
regulator of the cellular response to stress. Curr. Signal. Transduct. Ther. 5, 49-59.
5. Bond, M. R., and Hanover, J. A. (2013) O-GlcNAc Cycling: A Link Between
Metabolism and Chronic Disease. Annu. Rev. Nutr. 33, 205-229.
6. Okuyama, R., and Marshall, S. (2003) UDP-N-acetylglucosaminyl transferase (OGT) in
brain tissue: temperature sensitivity and subcellular distribution of cytosolic and
nuclear enzyme. J. Neurochem. 86, 1271-1280.
7. Griffith, L. S., and Schmitz, B. (1995) O-linked N-acetylglucosamine is upregulated in
Alzheimer brains. Biochem. Biophys. Res. Commun. 213, 424-431.
8. Arnold, C. S., Johnson, G. V., Cole, R. N., Dong, D. L., Lee, M., and Hart, G. W. (1996)
The microtubule-associated protein tau is extensively modified with O-linked N-
acetylglucosamine. J. Biol. Chem. 271, 28741-28744.
9. Cole, R. N., and Hart, G. W. (1999) Glycosylation sites flank phosphorylation sites on
synapsin I: O-linked N-acetylglucosamine residues are localized within domains
mediating synapsin I interactions. J. Neurochem. 73, 418-428.
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
11
10. Dong, D. L., Xu, Z. S., Hart, G. W., and Cleveland, D. W. (1996) Cytoplasmic O-
GlcNAc modification of the head domain and the KSP repeat motif of the
neurofilament protein neurofilament-H. J. Biol. Chem. 271, 20845-20852.
11. Liu, F., Shi, J., Tanimukai, H., Gu, J., Grundke-Iqbal, I., Iqbal, K., and Gong, C. X.
(2009) Reduced O-GlcNAcylation links lower brain glucose metabolism and tau
pathology in Alzheimer's disease. Brain 132, 1820-1832.
12. Mittal, S., and Ganesh, S. (2010) Protein quality control mechanisms and
neurodegenerative disorders: Checks, balances and deadlocks. Neurosci. Res. 68, 159-
166.
13. Zheng, Z., and Diamond, M.I. (2012) Huntington disease and the huntingtin protein.
Prog. Mol. Biol. Transl. Sci. 107, 189-214.
14. Cattaneo, E., Zuccato, C., and Tartari, M. (2005) Normal huntingtin function: an
alternative approach to Huntington's disease. Nat. Rev. Neurosci. 6, 919-930.
15. Ho, L. W., Brown, R., Maxwell, M., Wyttenbach, A., and Rubinsztein, D. C. (2001)
Wild type huntingtin reduces the cellular toxicity of mutant Huntingtin in mammalian
cell models of Huntington's disease. J. Med. Genet. 38, 450-452.
16. Rozas, J.L., Gómez-Sánchez, L., Tomás-Zapico, C., Lucas, J.J., and Fernández-Chacón,
R. (2011) Increased neurotransmitter release at the neuromuscular junction in a mouse
model of polyglutamine disease. J. Neurosci. 31, 1106-1113.
17. Shirendeb, U., Reddy, A. P., Manczak, M., Calkins, M. J., Mao, P., Tagle, D. A., and
Reddy, P. H. (2011) Abnormal mitochondrial dynamics, mitochondrial loss and mutant
huntingtin oligomers in Huntington's disease: implications for selective neuronal
damage. Hum. Mol. Genet. 20, 1438-1455.
18. Nucifora, F. C., Jr., Sasaki, M., Peters, M. F., Huang, H., Cooper, J. K., Yamada, M.,
Takahashi, H., Tsuji, S., Troncoso, J., Dawson, V. L., Dawson, T. M., and Ross, C. A.
(2001) Interference by huntingtin and atrophin-1 with cbp-mediated transcription
leading to cellular toxicity. Science 291, 2423-2428.
19. Ravikumar, B., Stewart, A., Kita, H., Kato, K., Duden, R., and Rubinsztein, D.C. (2003)
Raised intracellular glucose concentrations reduce aggregation and cell death caused by
mutant huntingtin exon 1 by decreasing mTOR phosphorylation and inducing
autophagy. Hum. Mol. Genet. 12, 985-994.
20. Puri, R., Jain, N., and Ganesh, S. (2011) Increased glucose concentration results in
reduced proteasomal activity and the formation of glycogen positive aggresomal
structures. FEBS J. 278, 3688-3698.
21. Kaniuk, N. A., Kiraly, M., Bates, H., Vranic, M., Volchuk, A., and Brumell, J. H. (2007)
Ubiquitinated-protein aggregates form in pancreatic beta-cells during diabetes-induced
oxidative stress and are regulated by autophagy. Diabetes 56, 930-939.
22. Cohen, E., and Dillin, A. (2008) The insulin paradox: aging, proteotoxicity and
neurodegeneration. Nat. Rev. Neurosci. 9, 759-767.
23. Steffan, J. S., Bodai, L., Pallos, J., Poelman, M., McCampbell, A., Apostol, B. L.,
Kazantsev, A., Schmidt, E., Zhu, Y. Z., Greenwald, M., Kurokawa, R., Housman, D.
E., Jackson, G. R., Marsh, J. L., and Thompson, L. M. (2001) Histone deacetylase
inhibitors arrest polyglutamine-dependent neurodegeneration in Drosophila. Nature
413, 739-743.
24. Freeman, M. (1996) Reiterative use of the EGF receptor triggers differentiation of all cell
types in the Drosophila eye. Cell 87, 651-60.
25. Dwivedi, V., Anandan, E. M., Mony, R. S., Muraleedharan, T. S., Valiathan, M. S.,
Mutsuddi, M., and Lakhotia, S. C. (2012) In vivo effects of traditional Ayurvedic
formulations in Drosophila melanogaster model relate with therapeutic applications.
PLoS One 7, e37113.
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
12
26. Garyali, P., Siwach, P., Singh, P.K., Puri, R., Mittal, S., Sengupta, S., Parihar, R., and
Ganesh, S. (2009) The malin-laforin complex suppresses the cellular toxicity of
misfolded proteins by promoting their degradation through the ubiquitin-proteasome
system. Hum. Mol. Genet. 18, 688-700.
27. Pampliega, O., Orhon, I., Patel, B., Sridhar, S., Díaz-Carretero, A., Beau, I., Codogno, P.,
Satir, B.H., Satir, P., and Cuervo, A.M. (2013) Functional interaction between
autophagy and ciliogenesis. Nature 502, 194-200.
28. Juenemann, K., Schipper-Krom, S., Wiemhoefer, A., Kloss, A., Sanz Sanz, A., and Reits,
E.A. (2013) Expanded polyglutamine-containing N-terminal huntingtin fragments are
entirely degraded by mammalian proteasomes. J. Biol. Chem. 288, 27068-27084.
29. Franceschini, N., and Kirschfeld, K. (1971) Pseudopupil phenomena in the compound
eye of drosophila. Kybernetik 9, 159-182.
30 Ravikumar, B., Sarkar, S., and Rubinsztein, D. C. (2008) Clearance of mutant aggregate-
prone proteins by autophagy. Methods Mol. Biol. 445, 195-211.
31 Matsumoto, G., Wada, K., Okuno, M., Kurosawa, M., and Nukina, N. (2011) Serine 403
phosphorylation of p62/SQSTM1 regulates selective autophagic clearance of
ubiquitinated proteins. Mol. Cell 44, 279-289.
32. Jensen, R. V., Zachara, N. E., Nielsen, P. H., Kimose, H. H., Kristiansen, S. B., and
Botker, H. E. (2013) Impact of O-GlcNAc on cardioprotection by remote ischaemic
preconditioning in non-diabetic and diabetic patients. Cardiovasc. Res. 97, 369-378.
33. Rajapakse, A. G., Ming, X. F., Carvas, J. M., and Yang, Z. (2009) The hexosamine
biosynthesis inhibitor azaserine prevents endothelial inflammation and dysfunction
under hyperglycemic condition through antioxidant effects. Am. J. Physiol. Heart Circ.
Physiol. 296, H815-822.
34. Mizushima, N., Yoshimori, T., and Levine, B. (2010) Methods in mammalian autophagy
research. Cell 140, 313-326.
35. Kimura, S., Noda, T., and Yoshimori, T. (2007) Dissection of the autophagosome
maturation process by a novel reporter protein, tandem fluorescent-tagged LC3.
Autophagy 3, 452-460.
36. Mallik, M., and Lakhotia, S. C. (2009) RNAi for the large non-coding hsromega
transcripts suppresses polyglutamine pathogenesis in Drosophila models. RNA Biol 6,
464-478.
37. Mallik, M., and Lakhotia, S. C. (2010) Modifiers and mechanisms of multi-system
polyglutamine neurodegenerative disorders: lessons from fly models. J. Genet. 89, 497-
526.
38. Jackson, G. R., Salecker, I., Dong, X., Yao, X., Arnheim, N., Faber, P. W., MacDonald,
M. E., and Zipursky, S. L. (1998) Polyglutamine-expanded human huntingtin
transgenes induce degeneration of Drosophila photoreceptor neurons. Neuron 21, 633-
642.
39. Akimoto, Y., Hart, G. W., Hirano, H., and Kawakami, H. (2005) O-GlcNAc
modification of nucleocytoplasmic proteins and diabetes. Med. Mol. Morphol. 38, 84-
91.
40. Alfaro, J. F., Gong, C. X., Monroe, M. E., Aldrich, J. T., Clauss, T. R., Purvine, S. O.,
Wang, Z., Camp, D. G., 2nd, Shabanowitz, J., Stanley, P., Hart, G. W., Hunt, D. F.,
Yang, F., and Smith, R. D. (2012) Tandem mass spectrometry identifies many mouse
brain O-GlcNAcylated proteins including EGF domain-specific O-GlcNAc transferase
targets. Proc. Natl. Acad. Sci. U S A. 109, 7280-7285.
41. Jadhav, S., Zilka, N., and Novak, M. (2013) Protein Truncation as a Common
Denominator of Human Neurodegenerative Foldopathies. Mol. Neurobiol. 48, 516-532.
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
13
42. Guinez, C., Mir, A. M., Dehennaut, V., Cacan, R., Harduin-Lepers, A., Michalski, J. C.,
and Lefebvre, T. (2008) Protein ubiquitination is modulated by O-GlcNAc
glycosylation. FASEB J. 22, 2901-2911.
43. Zhang, F., Su, K., Yang, X., Bowe, D. B., Paterson, A. J., and Kudlow, J. E. (2003) O-
GlcNAc modification is an endogenous inhibitor of the proteasome. Cell 115, 715-725.
44. Liu, K., Paterson, A. J., Zhang, F., McAndrew, J., Fukuchi, K., Wyss, J. M., Peng, L.,
Hu, Y., and Kudlow, J. E. (2004) Accumulation of protein O-GlcNAc modification
inhibits proteasomes in the brain and coincides with neuronal apoptosis in brain areas
with high O-GlcNAc metabolism. J. Neurochem. 89, 1044-1055.
45. Wang, P., Lazarus, B. D., Forsythe, M. E., Love, D. C., Krause, M. W., and Hanover, J.
A. (2012) O-GlcNAc cycling mutants modulate proteotoxicity in Caenorhabditis
elegans models of human neurodegenerative diseases. Proc. Natl. Acad. Sci. U S A 109,
17669-17674.
46. Bence, N. F., Sampat, R. M., and Kopito, R. R. (2001) Impairment of the ubiquitin-
proteasome system by protein aggregation. Science 292, 1552-1555.
47. Holmberg, C. I., Staniszewski, K. E., Mensah, K. N., Matouschek, A., and Morimoto, R.
I. (2004) Inefficient degradation of truncated polyglutamine proteins by the
proteasome. EMBO J. 23, 4307-4318.
48. Williams, A., Jahreiss, L., Sarkar, S., Saiki, S., Menzies, F. M., Ravikumar, B., and
Rubinsztein, D. C. (2006) Aggregate-prone proteins are cleared from the cytosol by
autophagy: therapeutic implications. Curr. Top. Dev. Biol. 76, 89-101.
49. Wells, L., Vosseller, K., and Hart, G. W. (2003) A role for N-acetylglucosamine as a
nutrient sensor and mediator of insulin resistance. Cell Mol. Life Sci. 60, 222-228.
50. Mizushima, N. (2007) Autophagy: process and function. Genes Dev. 21, 2861-2873.
51. Chang, Y. Y., Juhasz, G., Goraksha-Hicks, P., Arsham, A. M., Mallin, D. R., Muller, L.
K., and Neufeld, T. P. (2009) Nutrient-dependent regulation of autophagy through the
target of rapamycin pathway. Biochem. Soc. Trans. 37, 232-236.
52. Hull, R. L., Zraika, S., Udayasankar, J., Kisilevsky, R., Szarek, W. A., Wight, T. N., and
Kahn, S. E. (2007) Inhibition of glycosaminoglycan synthesis and protein glycosylation
with WAS-406 and azaserine result in reduced islet amyloid formation in vitro. Am. J.
Physiol. Cell Physiol. 293, C1586-1593.
53. Marsh, S. A., Powell, P. C., Dell'italia, L. J., and Chatham, J. C. (2013) Cardiac O-
GlcNAcylation blunts autophagic signaling in the diabetic heart. Life Sci. 92, 648-656.
Acknowledgments
We thank Dr Lawrence Marsh (University of California, Irvine, USA) for
the mHtt-Q97-GFP expression construct, Dr T. Yoshimori (National Institute for Basic
Biology, Okazaki, Japan) for the mRFP-GFP-LC3 construct, Dr John A Hannover (National
Institutes of Health, USA) and Dr Gerald W. Hart (Johns Hopkins University School of
Medicine, USA) for the expression constructs coding for OGA and OGT, respectively. We
would also like to thank the anonymous reviewers for their suggestions and comments which
greatly helped in improving the manuscript.
FOOTNOTES
*This work was supported by research grant from the Department of Atomic Energy (Govt.
of India) to SG. AK received a post-doctoral fellowship from the Department of
Biotechnology (Govt. of India). SG is a Ramanna Fellow and Gill-Joy Chair Professor at IIT
Kanpur and SCL is Professor Emeritus and a DAE-Raja Ramanna Fellow at Banaras Hindu
University, Varanasi.
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
14
4Current address: Burke Medical Research Institute, New York, USA.
5Current address: Institute of Genetics and Molecular and Cellular Biology, Strasbourg,
France.
6To whom correspondence should be addressed: Department of Biological Sciences and
Bioengineering, Indian Institute of Technology, Kanpur, 208016, India; Tel: (91) 512-259-
4040; Fax (91) 512-159-4010; Email: sganesh@iitk.ac.in
The abbreviations used are: DAPI, 4’,6-diamidino-2-phenylindole dihydrochloride; mHtt,
polyglutamine expanded huntingtin exon 1 protein fragment; OGT, O-linked N-
acetylglycosyl transferase; OGA, O-linked N-acetylglucosaminidase; PTM, post-translational
modification; HBP, hexosamine biosynthetic pathway; GFP, green fluorescence protein;
BafA1, bafilomycin A1; polyQ, polyglutamine; SDS, sodium dodecyl sulfate.
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
15
Figure 1: Suppression of O-GlcNAcylation significantly reduces mHtt-Q97 aggregates.
(A) Neuro2A cells transfected with an empty vector (pcDNA) or expression construct for
OGA-Myc and OGT-HA were evaluated for changes in global O-glycosylation level by
immunoblotting. Expression of OGA and OGT was confirmed by probing with the tag
antibodies. Probing with γ-tubulin served as loading control. The bar diagram shown above
represent the fold change in the signal intensity of O-glycosylated proteins (normalized to γ-
tubulin in the immunoblot) as measured by densitometric analysis (N=3; ***, p<0.001; *,
p<0.1). (B) Bar diagram representing percent transfected cells showing the aggregation of
mHtt-Q97-GFP when expressed alone (pcDNA) or with an expression construct coding for
OGA-Myc or OGT-HA, as indicated. Note the significant reduction in the transfected cells
positive for mHtt-Q97-GFP aggregates when OGA was co-expressed but a significant
increase in their frequency when OGT was co-expressed (N=3; ***, p<0.001). (C) Bar
diagram showing fold change in survival of cells transiently expressing OGA or OGT as
compared with cells transfected with an empty vector (pcDNA), as measured by MTT assay
(N=3; ***, p<0.001). (D) Representative fluorescence microscopic images (first four
columns with a 10X objective) showing aggregation patterns of mHtt-Q97-GFP in Neuro2A
cells when expressed alone (pcDNA), and when co-expressed with OGA-Myc or OGT-HA.
The intense green signals in “mHtt-Q97-GFP” column represent mHtt-Q97 aggregates. The
red signal reveals the expression of OGA-Myc or OGT-HA. Nuclei were stained with DAPI
(blue). Areas boxed in the “merged” column are enlarged in the last column to more clearly
show the GFP-positives cells with or without aggregates.
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
16
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
17
Figure 2: O-GlcNAcylation inhibition reduces mHtt-Q97-mediated cytotoxicity (A)
Western blot images of insoluble, aggregated form of mHtt-Q97-GFP in filter-trap assay
using a slot-blot apparatus (top) or its total form resolved by immunoblotting (bottom) when
expressed with either pcDNA (empty vector control), OGT or OGA, as indicated in middle.
Expression of OGT and OGA was established by probing them with anti-HA and anti-Myc
antibodies, respectively levels. The bar diagram above shows the fold changes in signal
intensities, based on densitometric analysis, of SDS insoluble, aggregated form of mHtt-Q97-
GFP (normalized to total level detected in the immunoblot; N=3; ***, p<0.001; *, p<0.1).
(B, C) Bar diagram representing the fold change in the viability of cells expressing mHtt-
Q97-GFP (B) or the α-synuclein mutant A40P (C) as measured by an MTT assay. Cells
transfected with indicated constructs were processed for the measurement, and in each set the
value obtained for the GFP transfected cells was considered and as 1, and the relative values
obtained for indicated combinations were plotted. (D, E) Bar diagram showing the percentage
of cells expressing mHtt-Q97-GFP (D) or the α-synuclein mutant A40P (E) with abnormal
(apoptotic) nuclei (as shown in F) as compared with cells that expressed GFP (control) when
co-transfected with OGA or OGT coding constructs (in B-E, N=3; **, P<0.05; ***, P<0.005
on Student's t-test). (F) Representative images showing a normal (left) and an abnormal
(apoptotic; right) nuclei as judged by DPAI staining (scale, 5 µM).
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
18
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
19
Figure 3: O-GlcNAcylation modulates autophagy. (A) Immunoblots (bottom panel) of
Neuro2A cells, transiently expressing pcDNA empty vector alone or OGA-Myc or OGT-HA
for 36 hrs, to show levels of the autophagic markers, LC3II and p62. Probing with anti-γ-
tubulin served as loading control. Note the change in the level of LC3II band (identified by
an arrow) in cells that expressed OGA. Co-expression of OGT did not show such an effect.
Bar diagrams above show the fold changes in signal intensities of the LC3II and p62 (both
normalized to γ-tubulin signal) bands when compared with the control (pcDNA transfected
cells). (B) Neuro2A cells were grown in a medium with or without azaserine and or
glucosamine for 12 hrs as indicated, and the changes in the global glycosylation level were
evaluated. The bar diagrams above show the fold changes in the glycosylation levels
compared to cells that were fed with glucose. (C) Samples shown in B were tested for the
level of autophagy markers LC3 and p62 as indicated. Note the reduction in the intensity of
the band for LC3II (identified by an arrow) and p62 in the azaserine treated cells and their
restoration in the azaserine/glucosamine double treated cells. Bar diagrams above represent
the fold changes in the signal intensities for LC3II and p62 (both normalized to γ-tubulin
signal) bands compared to the control (glucose fed cells) (in A-C, N=3; *, P<0.5; **, P<0.05;
***, P<0.005 on Student's t-test).
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
20
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
21
Figure 4: Suppression of O-GlcNAcylation increases autophagy flux. (A) Neuro2A cells
at 24 h post- transient transfection with an empty vector (pcDNA) or with a construct coding
for OGA or OGT were either left untreated or treated with BafA1 for 12 h as indicated and
the levels of autophagy markers LC3 and p62 were evaluated by immunoblotting. Note the
increase in the signal intensities of LC3II (arrow) and p62 in all samples treated with BafA1.
The blot was probed with anti-Myc and Anti-HA antibodies to show the expression of OGA
and OGT, respectively; probing with anti-γ-tubulin served as the loading a control. (B)
Immunoblot to show levels of LC3II (arrow) and p62 in Neuro2A cells, as in A, untreated or
treated with azaserine, alone or in combination with BafA1 as indicated; γ-ubulin served as
loading control. Bar diagrams above represent the fold changes in the signal intensities for
LC3II and p62 (both normalized to γ-tubulin signal) bands compared to the control (N=3; **,
P<0.05; ***, P<0.005 on Student's t-test).
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
22
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
23
Figure 5: Suppression of O-GlcNAcylation increases autophagy flux. (A) Representative
images of cells showing LC3 positive puncta in cells that were transiently transfected with
mRFP–GFP–LC3 expression construct along with an empty vector (pcDNA) or an
expression construct coding for OGA or OGT as indicated. Puncta that are positive both for
red and green fluorescence represent autophagosomes while those positive only for red
represent autolysosomes (bar = 10 µM). (B) Bar diagram showing the fraction of puncta
positive for both RFP and GFP (yellow) or only the RFP (red) in transiently transfected cells
coexpressing mRFP–GFP–LC3 and pcDNA or OGA or OGT, as indicated. N=3; *, P<0.5;
**, P<0.05 on Student's t-test).
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
24
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
25
Figure 6: Suppression of O-GlcNAcylation increases autophagy flux. Western blots
showing changes in the levels of insoluble, aggregated form of mHtt-Q97-GFP (filter-trap
assay; top) or its total form (immunoblotting; bottom) when expressed with OGA and treated
or not treated with BafA1 (A) or 3-MA (B) as indicated. The bar diagrams, shown above,
represent fold changes in the signal intensity of SDS insoluble, aggregated form of mHtt-
Q97-GFP (normalized to total level detected in the immunoblot) as measured by
densitometric analysis (N=3; ***, p<0.001; *, p<0.1).
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
26
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
27
Figure 7: Effect of O-GlcNAcylation on proteasomal activity. Bar diagram showing fold
change in the proteasomal activity in cells transiently transfected with a construct coding for
OGA, OGT or an empty vector (pcDAN) (A) or with the drug azaserine or glucosamine in
the presence or absence proteasomal blocker MG132, as indicated (N=3; *, P<0.5; **,
P<0.05 on Student's t-test).
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
28
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
29
Figure 8: Azaserine feeding reduces accumulation of mutant Huntingtin protein in fly
model. (A-D) Confocal projection images (projections of 4 consecutive optical sections
which show the morphogenetic furrow) of eye imaginal discs of late 3rd instar GMR-
GAL4>UAS-httex1p Q93 Drosophila larvae, reared from the 1st instar stage onwards on
normal (A, B) or azaserine supplemented food (C, D), immunostained for HA-tagged mutant
Htt (green, A-D, identified as “PolyQ”); nuclei are counterstained with DAPI (blue, B and D).
The insets in A and C are higher magnification images of a part of the eye discs in A and C,
respectively, to more clearly show the polyQ aggregates, which are very abundant in A but
nearly absent in C. Arrows in B and D indicate position of the morphogenetic furrow. Scale
bar in A represents 20 μm and applies to A to D. (E) Immunoblot of total proteins from heads
of one day old GMR-GAL4/UAS-htt-ex1p Q93 flies, reared on normal (Aza -) or azaserine
supplemented (Aza +) food since the 1st instar stage, probed with anti-HA antibody to detect
Htt-Q93 protein. (F) Histograms show mean relative levels of HA-tagged polyQ protein
(mean ratios of Htt-Q93 and γ-tubulin densities) determined from triplicate immunoblots as
in E; the mean ratio of HttQ93 and γ-tubulin densities in Aza- food was taken as 1. (*
p<0.001).
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
30
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
31
Figure 9: Azaserine feeding suppresses mHttQ93-induced neurodegeneration in adult
Drosophila eyes and reduces the age-dependent loss of vision. (A-C) Pseudopupil images
of eyes of 1 day old wild type (A), or GMR-GAL4>UAS-httex1p Q93 flies grown on control
(B) or on azaserine-containing food (C). Arrow in C indicates presence of two distinct
rhabdomeres in one of the ommatidial units; these are not seen in any ommatidial unit in
control flies. Scale bar in A indicates 20 μm and applies to A C. (D) Histograms showing
phototaxis (percent flies moving to illuminated chamber, Y-axis) of wild type and GMR-
GAL4>UAS-httex1p Q93 flies reared on control or azaserine supplemented food on different
days (X – axis) after emergence. Each value in bar diagram is mean of 20 replicates with 10
flies in each set. The * in bar diagrams indicates the p-value to be <0.05when comparing the
mean phototaxis of GMR-GAL4>UAS-httex1p Q93 flies reared on control and azaserine
supplemented food, respectively, on days 5, 10 and 15 .
by guest on March 20, 2014http://www.jbc.org/Downloaded from
Decreasing O-linked GlcNAcylation increases basal autophagic flux
32
by guest on March 20, 2014http://www.jbc.org/Downloaded from
... However, mHTT protein is intracellularly cleared mainly through autophagy-lysosome pathway, and O-GlcNAcylation mutually inhibits autophagy. Due to these facts, OGT-induced O-GlcNAcylation suppressed autophagic degradation of mHTT protein and enhanced toxicity (Kumar et al., 2014). on the other hand, downregulation of O-GlcNAcylation enhanced the viability of neuronal cells expressing mHtt. ...
Article
Artemisinin (ART) and its derivatives, collectively referred to as artemisinins (ARTs), have been approved for the treatment of malaria for decades. ARTs are converted into dihydroartemisinin (DHA), the only active form, which is reductive in vivo. In this review, we provide a brief overview of the neuroprotective potential of ARTs and the underlying mechanisms on several of the most common neurodegenerative diseases, particularly considering their potential application in those associated with cognitive and motor impairments including Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), and amyotrophic lateral sclerosis (ALS). ARTs act as autophagy balancers to alleviate AD and PD. They inhibit neuroinflammatory responses by regulating phosphorylation of signal transduction proteins, such as AKT, PI3K, ERK, NF-[Formula: see text]B, p38 MAPK, I[Formula: see text]B[Formula: see text]. In addition, ARTs regulate GABAergic signaling in a dose-dependent manner. Although they competitively inhibit the binding of gephyrin to GABAergic receptors, low doses of ARTs enhance GABAergic signaling. ARTs can also inhibit ferroptosis, activate the Akt/Bcl-2, AMPK, or ERK/CREB pathways to reduce oxidative stress, and maintain mitochondrial homeostasis, protecting neurons from oxidative stress injury. More importantly, ARTs structurally combine with and suppress [Formula: see text]-Amyloid (A[Formula: see text]-induced neurotoxicity, reduce P-tau, and maintain O-GlcNAcylation/Phosphorylation balance, leading to relieved pathological changes in neurodegenerative diseases. Collectively, these natural properties endow ARTs with unique potential for application in neurodegenerative diseases.
... O-linked β-Nacetylglucosamine (O-GlcNAc) modification of the proteasomal 19S regulatory cap Rpt2 protein and S-glutathionylation of the proteasomal 19S regulatory cap RPN1 and RPN2 proteins are reported to reduce proteasome activity and lead to the accumulation of ubiquitylated protein (Zhang et al., 2003;Zmijewski et al., 2009;Keembiyehetty et al., 2011;Liu et al., 2014). Interestingly, upregulated O-GlcNAcylation and S-glutathionylation modified proteins have been reported to impair cell homeostasis and contribute to neuron loss in HD model mice and model cells (Yuzwa et al., 2008;Kumar et al., 2014;Hong et al., 2015;Grima et al., 2017;Lee et al., 2021). Therefore, oleuropein may modulate O-GlcNAcylation or S-glutathionylation modification of the proteasomal proteins to improve proteasome activity and relieve oxidative stress of the mHtt-expressing cells. ...
Article
Full-text available
Introduction Huntington’s disease (HD) is a hereditary neurodegenerative disorder that primarily affects the striatum, a brain region responsible for movement control. The disease is characterized by the mutant huntingtin (mHtt) proteins with an extended polyQ stretch, which are prone to aggregation. These mHtt aggregates accumulate in neurons and are the primary cause of the neuropathology associated with HD. To date, no effective cure for HD has been developed. Methods The immortalized STHdh Q111/Q111 striatal cell line, the mHtt-transfected wild-type STHdh Q7/Q7 striatal cell line, and N2a cells were used as Huntington's disease cell models. Flow cytometry was used to assess cellular reactive oxygen species and transfection efficiency. The CCK-8 assay was used to measure cell viability, while fluorescence microscopy was used to quantify aggregates. Immunoblotting analyses were used to evaluate the effects on protein expression. Results Polyphenols are natural antioxidants that offer neuroprotection in neurological disorders. In this study, we provide evidence that oleuropein, the primary polyphenol in olive leaves and olive oil, enhances cell viability in HD cell models, including. STHdh Q7/Q7 STHdh Q7/Q7 striatal cells, N2a cells ectopically expressing the truncated mHtt, and STHdh Q111/Q111 striatal cells expressing the full-length mHtt. Oleuropein effectively reduced both soluble and aggregated forms of mHtt protein in these HD model cells. Notably, the reduction of mHtt aggregates associated with oleuropein was linked to increased proteasome activity rather than changes in autophagic flux. Oleuropein seems to modulate proteasome activity through an unidentified pathway, as it did not affect the 20S proteasome catalytic β subunits, the proteasome regulator PA28γ, or multiple MAPK pathways. Discussion We demonstrated that oleuropein enhances the degradation of mHtt by increasing proteasomal protease activities and alleviates mHtt-induced cytotoxicity. Hence, we propose that oleuropein and potentially other polyphenols hold promise as a candidate for alleviating Huntington's disease.
... However, the higher content of Cu or persistent organic pollutants such as polycyclic aromatic hydrocarbons (PAHs), found in the aerosols, could have the potential to inhibit phytoplankton growth (Jordi et al., 2012;Castro-Jimenez et al., 2012;Berrojalbiz et al., 2014), due to their toxicity at high concentrations (Paytan et al., 2009). The concept of toxicity is currently controversial since the concentration of trace metals in seawater due to atmospheric deposition is much lower than the traditionally toxic threshold that causes obvious cell death (Kumar et al., 2014;Paytan et al., 2009). Finally, the questions that still had to be addressed were related to the identification of trace metals critical for phytoplankton growth, and also to the degree of their bioavailability which depend on their fractional solubility, i.e. the dissolved fraction of this element in contact with seawater (Richon et al., 2018). ...
... Additionally, the results involving the p62 levels supported the hypothesis that TMG induces autophagy by decreasing p62 levels, whereas Osmi-1 inhibited autophagosome-lysosome fusion, leading to increased p62 levels. Previous works have demonstrated that both increases [66] and reductions in O-GlcNAcylation levels can contribute to autophagy [29,68]. Indeed, different works have already demonstrated that autophagy favors the survival and therapeutic resistance of GB [28,[69][70][71]. ...
Article
Full-text available
Simple Summary Glioblastoma (GB) is the most common and aggressive type of malignant brain tumor; however, despite advances in treatment modalities, it remains largely incurable. Current therapeutic protocols are ineffective, and temozolomide (TMZ), the main chemotherapy used in GB treatment, has a high resistance rate. Aberrant O-GlcNAcylation is related to the tumorigenesis of several tumor types, and targeting O-GlcNAc transferase (OGT) is a possible therapeutic target for some tumor types. Here, we investigated the effect of OGT inhibition on cellular proliferation, cell death, and autophagy, as well as whether it could improve the effect of TMZ on cell viability. Our findings indicated that targeting OGT shows promising potential as a therapeutic strategy for treating GB. Abstract Glioblastoma (GB) is the most aggressive primary malignant brain tumor and is associated with short survival. O-GlcNAcylation is an intracellular glycosylation that regulates protein function, enzymatic activity, protein stability, and subcellular localization. Aberrant O-GlcNAcylation is related to the tumorigenesis of different tumors, and mounting evidence supports O-GlcNAc transferase (OGT) as a potential therapeutic target. Here, we used two human GB cell lines alongside primary human astrocytes as a non-tumoral control to investigate the role of O-GlcNAcylation in cell proliferation, cell cycle, autophagy, and cell death. We observed that hyper O-GlcNAcylation promoted increased cellular proliferation, independent of alterations in the cell cycle, through the activation of autophagy. On the other hand, hypo O-GlcNAcylation inhibited autophagy, promoted cell death by apoptosis, and reduced cell proliferation. In addition, the decrease in O-GlcNAcylation sensitized GB cells to the chemotherapeutic temozolomide (TMZ) without affecting human astrocytes. Combined, these results indicated a role for O-GlcNAcylation in governing cell proliferation, autophagy, cell death, and TMZ response, thereby indicating possible therapeutic implications for treating GB. These findings pave the way for further research and the development of novel treatment approaches which may contribute to improved outcomes and increased survival rates for patients facing this challenging disease.
Article
Full-text available
O-GlcNAcylation, a dynamic post-translational modification occurring on serine or threonine residues of numerous proteins, plays apivotal role in various cellular processes, including gene regulation, metabolism, and stress response. Abundant in the brain,O-GlcNAcylation intricately governs neurodevelopment, synaptic assembly, and neuronal functions. Recent investigations haveestablished a correlation between the dysregulation of brain O-GlcNAcylation and a broad spectrum of neurological disorders andinjuries, spanning neurodevelopmental, neurodegenerative, and psychiatric conditions, as well as injuries to the central nervoussystem (CNS). Manipulating O-GlcNAcylation has demonstrated neuroprotective properties against these afflictions. This reviewdelineates the roles and mechanisms of O-GlcNAcylation in the CNS under both physiological and pathological circumstances, witha focus on its neuroprotective effects in neurological disorders and injuries. We discuss the involvement of O-GlcNAcylation in keyprocesses such as neurogenesis, synaptic plasticity, and energy metabolism, as well as its implications in conditions like Alzheimer’sdisease, Parkinson’s disease, and ischemic stroke. Additionally, we explore prospective therapeutic approaches for CNS disordersand injuries by targeting O-GlcNAcylation, highlighting recent clinical developments and future research directions. Thiscomprehensive overview aims to provide insights into the potential of O-GlcNAcylation as a therapeutic target and guide futureinvestigations in this promising field.
Article
Full-text available
As a non-classical post-translational modification, O-linked β-N-acetylglucosamine (O-GlcNAc) modification (O-GlcNAcylation) is widely found in human organ systems, particularly in our brains, and is indispensable for healthy cell biology. With the increasing age of the global population, the incidence of neurodegenerative diseases is increasing, too. The common characteristic of these disorders is the aggregation of abnormal proteins in the brain. Current research has found that O-GlcNAcylation dysregulation is involved in misfolding or aggregation of these abnormal proteins to mediate disease progression, but the specific mechanism has not been defined. This paper reviews recent studies on O-GlcNAcylation’s roles in several neurodegenerative disorders such as Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, Huntington’s disease, Machado-Joseph’s disease, and giant axonal neuropathy, and shows that O-GlcNAcylation, as glucose metabolism sensor, mediating synaptic function, participating in oxidative stress response and signaling pathway conduction, directly or indirectly regulates characteristic pathological protein toxicity and affects disease progression. The existing results suggest that targeting O-GlcNAcylation will provide new ideas for clinical diagnosis, prevention, and treatment of neurodegenerative diseases.
Article
O-GlcNAc is a common modification found on nuclear and cytoplasmic proteins. Determining the catalytic mechanism of the enzyme O-GlcNAcase (OGA), which removes O-GlcNAc from proteins, enabled the creation of potent and selective inhibitors of this regulatory enzyme. Such inhibitors have served as important tools in helping to uncover the cellular and organismal physiological roles of this modification. In addition, OGA inhibitors have been important for defining the augmentation of O-GlcNAc as a promising disease-modifying approach to combat several neurodegenerative diseases including both Alzheimer’s disease and Parkinson’s disease. These studies have led to development and optimization of OGA inhibitors for clinical application. These compounds have been shown to be well tolerated in early clinical studies and are steadily advancing into the clinic. Despite these advances, the mechanisms by which O-GlcNAc protects against these various types of neurodegeneration are a topic of continuing interest since improved insight may enable the creation of more targeted strategies to modulate O-GlcNAc for therapeutic benefit. Relevant pathways on which O-GlcNAc has been found to exert beneficial effects include autophagy, necroptosis, and processing of the amyloid precursor protein. More recently, the development and application of chemical methods enabling the synthesis of homogenous proteins have clarified the biochemical effects of O-GlcNAc on protein aggregation and uncovered new roles for O-GlcNAc in heat shock response. Here, we discuss the features of O-GlcNAc in neurodegenerative diseases, the application of inhibitors to identify the roles of this modification, and the biochemical effects of O-GlcNAc on proteins and pathways associated with neurodegeneration.
Article
Full-text available
Under normal physiological conditions, the mammalian brain contains very little glycogen, most of which is stored in astrocytes. However, the aging brain and subareas of the brain in patients with neurodegenerative disorders tend to accumulate glycogen, the cause and significance of which remain largely unexplored. Using cellular models, we have recently demonstrated a neuroprotective role for neuronal glycogen and glycogen synthase in the context of Huntington's disease. To gain insight into the role of brain glycogen in regulating proteotoxicity, we utilized a Drosophila model of Huntington's disease in which glycogen synthase was either knocked down or expressed ectopically. Enhancing glycogen synthesis in the brains of flies with Huntington's disease decreased mutant Huntingtin aggregation and reduced oxidative stress by activating auto-lysosomal functions. Further, the overexpression of glycogen synthase in the brain rescues photoreceptor degeneration, improves locomotor deficits, and increases fitness traits in Huntington's disease model. We thus provide in vivo evidence for the neuroprotective functions of glycogen synthase and glycogen in neurodegenerative conditions and their role in the neuronal autophagy process.
Article
Full-text available
Nutrient deprivation is a stimulus shared by both autophagy and the formation of primary cilia. The recently discovered role of primary cilia in nutrient sensing and signalling motivated us to explore the possible functional interactions between this signalling hub and autophagy. Here we show that part of the molecular machinery involved in ciliogenesis also participates in the early steps of the autophagic process. Signalling from the cilia, such as that from the Hedgehog pathway, induces autophagy by acting directly on essential autophagy-related proteins strategically located in the base of the cilium by ciliary trafficking proteins. Whereas abrogation of ciliogenesis partially inhibits autophagy, blockage of autophagy enhances primary cilia growth and cilia-associated signalling during normal nutritional conditions. We propose that basal autophagy regulates ciliary growth through the degradation of proteins required for intraflagellar transport. Compromised ability to activate the autophagic response may underlie some common ciliopathies.
Article
Full-text available
Huntington disease is a neurodegenerative disorder caused by an expanded polyglutamine (polyQ) repeat within the protein huntingtin (Htt). N-terminal fragments of the mutant Htt (mHtt) proteins containing the polyQ repeat are aggregation-prone and form intracellular inclusion bodies. Improving the clearance of mHtt fragments by intracellular degradation pathways is relevant to obviate toxic mHtt species and subsequent neurodegeneration. Because the proteasomal degradation pathway has been the subject of controversy regarding the processing of expanded polyQ repeats, we examined whether the proteasome can efficiently degrade Htt-exon1 with an expanded polyQ stretch both in neuronal cells and in vitro. Upon targeting mHtt-exon1 to the proteasome, rapid and complete clearance of mHtt-exon1 was observed. Proteasomal degradation of mHtt-exon1 was devoid of polyQ peptides as partial cleavage products by incomplete proteolysis, indicating that mammalian proteasomes are capable of efficiently degrading expanded polyQ sequences without an inhibitory effect on the proteasomal activity.
Article
Full-text available
Neurodegenerative foldopathies are characterized by aberrant folding of diseased modified proteins, which are major constituents of the intracellular and extracellular lesions. These lesions correlate with the cognitive and/or motor impairment seen in these diseases. The majority of the disease modified proteins in neurodegenerative foldopathies belongs to the group of proteins termed as intrinsically disordered proteins (IDPs). Several independent studies have showed that abnormal protein processing constitutes the key pathological feature of these disorders. The current review focuses on protein truncation as a common denominator of neurodegenerative foldopathies, which is considered to be the major driving force behind the pathological metamorphosis of brain IDPs. The aim of the review is to emphasize the key role of the protein truncation in the pathogenic pathways of neurodegenerative diseases. A deeper understanding of the complex downstream processing of the IDPs, resulting in the generation of pathologically modified proteins might be a prerequisite for the successful therapeutic strategies of several fatal neurodegenerative diseases.
Article
Full-text available
Expanded polyglutamine repeats have been proposed to cause neuronal degeneration in Huntington's disease (HD) and related disorders, through abnormal interactions with other proteins containing short polyglutamine tracts such as the transcriptional coactivator CREB binding protein, CBP. We found that CBP was depleted from its normal nuclear location and was present in polyglutamine aggregates in HD cell culture models, HD transgenic mice, and human HD postmortem brain. Expanded polyglutamine repeats specifically interfere with CBP-activated gene transcription, and overexpression of CBP rescued polyglutamine-induced neuronal toxicity. Thus, polyglutamine-mediated interference with CBP-regulated gene transcription may constitute a genetic gain of function, underlying the pathogenesis of polyglutamine disorders.
Article
Full-text available
Aims: Post-translational modification of proteins by O-linked β-N-acetylglucosamine (O-GlcNAc) is cardioprotective but its role in cardioprotection by remote ischaemic preconditioning (rIPC) and the reduced efficacy of rIPC in type 2 diabetes mellitus is unknown. In this study we achieved mechanistic insight into the remote stimulus mediating and the target organ response eliciting the cardioprotective effect by rIPC in non-diabetic and diabetic myocardium and the influence of O-GlcNAcylation. Methods and results: The cardioprotective capacity and the influence on myocardial O-GlcNAc levels of plasma dialysate from eight healthy volunteers and eight type 2 diabetic patients drawn before and after subjection to an rIPC stimulus were tested on human isolated atrial trabeculae subjected to ischaemia/reperfusion injury. Dialysate from healthy volunteers exposed to rIPC improved post-ischaemic haemodynamic recovery (40 ± 6 vs. 16 ± 2%; P < 0.01) and increased myocardial O-GlcNAc levels. Similar observations were made with dialysate from diabetic patients before exposure to rIPC (43 ± 3 vs. 16 ± 2%; P < 0.001) but no additional cardioprotection or further increase in O-GlcNAc levels was achieved by perfusion with dialysate after exposure to rIPC (44 ± 4 and 42 ± 5 vs. 43 ± 3%; P = 0.7). The glutamine:fructose-6-phosphate amidotransferase (GFAT) inhibitor azaserine abolished the cardioprotective effects and the increment in myocardial O-GlcNAc levels afforded by plasma from diabetic patients and healthy volunteers treated with rIPC. Conclusions: rIPC and diabetes mellitus per se influence myocardial O-GlcNAc levels through circulating humoral factors. O-GlcNAc signalling participates in mediating rIPC-induced cardioprotection and maintaining a state of inherent chronic activation of cardioprotection in diabetic myocardium, restricting it from further protection by rIPC.
Article
Huntington's disease is caused by a CAG trinucleotide repeat expansion that is translated into an abnormally long polyglutamine tract. This gain-of-function mutation is associated with huntingtin aggregation and cell death. Autophagy is an important clearance route for mutant huntingtin exon 1. While mammalian target of rapamycin (mTOR) is a key regulator of autophagy, the upstream modifiers of this process are poorly understood. Our previous expression profiling studies in HD cell models observed changes in four genes associated with glucose metabolism, including the GLUT1 glucose transporter. A role for intracellular glucose as a modulator for polyglutamine toxicity was suggested as cell death was reduced by GLUT1 overexpression. Here we show that the protective effect of GLUT1 is associated with decreased huntingtin exon 1 aggregation in cell models. Consistent with this result, we also observed reduced aggregation and enhanced clearance of mutant huntingtin when cells were cultured in raised glucose concentrations (8 g/l). These effects were mimicked by 8 g/l 2-deoxyglucose (2DOG) (transported, phosphorylated but not metabolized further), but not with 8 g/l 3-O-methyl glucose (transported but not metabolized further). Thus, this phenomenon is probably mediated by glucose-6-phosphate. Increased clearance of mutant huntingtin by raised glucose (8 g/l) and 2DOG correlated with increased autophagy and reduced phosphorylation of mTOR, S6K1 and Akt. Thus, raised intracellular glucose/glucose 6-phosphate levels reduce mutant huntingtin toxicity by increasing autophagy via mTOR and possibly Akt. As mTOR and Akt regulate a diversity of crucial cellular processes, our data also suggest a major new set of targets for intracellular glucose signalling.
Article
Synapsin I is concentrated in nerve terminals, where it appears to anchor synaptic vesicles to the cytoskeleton and thereby ensures a steady supply of fusion-competent synaptic vesicles. Although phosphorylation-dependent binding of synapsin I to cytoskeletal elements and synaptic vesicles is well characterized, little is known about synapsin I’s O-linked N-acetylglucosamine (O-GlcNAc) modifications. Here, we identified seven in vivo O-GlcNAcylation sites on synapsin I by analysis of HPLC-purified digests of rat brain synapsin I. The seven O-GlcNAcylation sites (Ser55, Thr56, Thr87, Ser516, Thr524, Thr562, and Ser576) in synapsin I are clustered around its five phosphorylation sites in domains B and D. The proximity of phosphorylation sites to O-GlcNAcylation sites in the regulatory domains of synapsin I suggests that O-GlcNAcylation may modulate phosphorylation and indirectly affect synapsin I interactions. With use of synthetic peptides, however, the presence of an O-GlcNAc at sites Thr562 and Ser576 resulted in only a 66% increase in the Km of calcium/calmodulin-dependent protein kinase II phosphorylation of site Ser566 with no effect on its Vmax. We conclude that O-GlcNAcylation likely plays a more direct role in synapsin I interactions than simply modulating the protein’s phosphorylation.
Article
OBJECTIVES Recent data suggest that wild type huntingtin can protect against apoptosis in the testis of mice expressing full length huntingtin transgenes with expanded CAG repeats. It is not clear if this protective effect was confined to particular cell types, or if wild type huntingtin exerted its protective effect in this model by simply reducing the formation of toxic proteolytic fragments from mutant huntingtin. METHODS We cotransfected neuronal (SK-N-SH, human neuroblastoma) and non-neuronal (COS-7, monkey kidney) cell lines with HD exon 1 (containing either 21 or 72 CAG repeats) construct DNA and either full length wild type huntingtin or pFLAG (control vector). RESULTS Full length wild type huntingtin significantly reduced cell death resulting from the mutantHD exon 1 fragments containing 72 CAG repeats in both cell lines. Wild type huntingtin did not significantly modulate cell death caused by transfection of HD exon 1 fragments containing 21 CAG repeats in either cell line. CONCLUSIONS Our results suggest that wild type huntingtin can significantly reduce the cellular toxicity of mutant HD exon 1 fragments in both neuronal and non-neuronal cell lines. This suggests that wild type huntingtin can be protective in different cell types and that it can act against the toxicity caused by a mutant huntingtin fragment as well as against a full length transgene.
Article
To maintain homeostasis under variable nutrient conditions, cells rapidly and robustly respond to fluctuations through adaptable signaling networks. Evidence suggests that the O-linked N-acetylglucosamine (O-GlcNAc) posttranslational modification of serine and threonine residues functions as a critical regulator of intracellular signaling cascades in response to nutrient changes. O-GlcNAc is a highly regulated, reversible modification poised to integrate metabolic signals and acts to influence many cellular processes, including cellular signaling, protein stability, and transcription. This review describes the role O-GlcNAc plays in governing both integrated cellular processes and the activity of individual proteins in response to nutrient levels. Moreover, we discuss the ways in which cellular changes in O-GlcNAc status may be linked to chronic diseases such as type 2 diabetes, neurodegeneration, and cancers, providing a unique window through which to identify and treat disease conditions. Expected final online publication date for the Annual Review of Nutrition Volume 33 is July 17, 2013. Please see http://www.annualreviews.org/catalog/pubdates.aspx for revised estimates.
Article
The addition of N-acetylglucosamine (GlcNAc) O-linked to serine and threonine residues of proteins is known as O-GlcNAc. This post-translational modification is found within multicellular eukaryotes on hundreds of nuclear and cytoplasmic proteins. O-GlcNAc transferase (OGT) installs O-GlcNAc onto target proteins and O-GlcNAcase (OGA) removes O-GlcNAc. Their combined action makes O-GlcNAc reversible and serves to regulate cellular O-GlcNAc levels. Here I review select recent literature on the catalytic mechanism of these enzymes and studies on the molecular basis by which these enzymes identify and process their substrates. Molecular level understanding of how these enzymes work, and the basis for their specificity, should aid understanding how O-GlcNAc contributes to diverse cellular processes ranging from cellular signaling through to transcriptional regulation.