ArticlePDF AvailableLiterature Review

Abstract and Figures

Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by the expansion of a polyglutamine stretch within the huntingtin protein (HTT). The neurological symptoms, that involve motor, cognitive and psychiatric disturbances, are caused by neurodegeneration that is particularly widespread in the basal ganglia and cereberal cortex. HTT is ubiquitously expressed and in recent years it has become apparent that HD patients experience a wide array of peripheral organ dysfunction including severe metabolic phenotype, weight loss, HD-related cardiomyopathy and skeletal muscle wasting. Although skeletal muscles pathology became a hallmark of HD, the mechanisms underlying muscular atrophy in this disorder are unknown. Skeletal muscles account for approximately 40% of body mass and are highly adaptive to physiological and pathological conditions that may result in muscle hypertrophy (due to increased mechanical load) or atrophy (inactivity, chronic disease states). The atrophy is caused by degeneration of myofibers and their replacement by fibrotic tissue is the major pathological feature in many genetic muscle disorders. Under normal physiological conditions the muscle function is orchestrated by a network of intrinsic hypertrophic and atrophic signals linked to the functional properties of the motor units that are likely to be imbalanced in HD. In this article, we highlight the emerging field of research with particular focus on the recent studies of the skeletal muscle pathology and the identification of new disease-modifying treatments.
This content is subject to copyright.
MINI REVIEW ARTICLE
published: 06 October 2014
doi: 10.3389/fphys.2014.00380
Skeletal muscle pathology in Huntington’s disease
Daniel Zielonka1*, Izabela Piotrowska2, Jerzy T. Marcinkowski1and Michal Mielcarek3*
1Department of Social Medicine, Poznan University of Medical Sciences, Poznan, Poland
2MRC National Institute for Medical Research, London, UK
3Department of Medical and Molecular Genetics, King’s College London, London, UK
Edited by:
Julio L. Vergara, University of
California, Los Angeles, USA
Reviewed by:
Seth L. Robia, Loyola University
Chicago, USA
Andrew Alvin Voss, Wright State
University, USA
*Correspondence:
Daniel Zielonka, Department of
Social Medicine, Poznan University
of Medical Sciences, Rokietnicka
Str., No. 5 “C, 60-806 Poznan,
Poland
Michal Mielcarek, Department of
Medical and Molecular Genetics,
School of Medicine, King’s College
London, 8th Floor Tower Wing,
Guy’s Hospital Great Maze Pond,
London, SE1 9RT, UK
e-mail: michal.mielcarek@kcl.ac.uk
Huntington’s disease (HD) is a hereditary neurodegenerative disorder caused by the
expansion of a polyglutamine stretch within the huntingtin protein (HTT). The neurological
symptoms, that involve motor, cognitive and psychiatric disturbances, are caused by
neurodegeneration that is particularly widespread in the basal ganglia and cereberal
cortex. HTT is ubiquitously expressed and in recent years it has become apparent
that HD patients experience a wide array of peripheral organ dysfunction including
severe metabolic phenotype, weight loss, HD-related cardiomyopathy and skeletal muscle
wasting. Although skeletal muscles pathology became a hallmark of HD, the mechanisms
underlying muscular atrophy in this disorder are unknown. Skeletal muscles account for
approximately 40% of body mass and are highly adaptive to physiological and pathological
conditions that may result in muscle hypertrophy (due to increased mechanical load) or
atrophy (inactivity, chronic disease states). The atrophy is caused by degeneration of
myofibers and their replacement by fibrotic tissue is the major pathological feature in many
genetic muscle disorders. Under normal physiological conditions the muscle function is
orchestrated by a network of intrinsic hypertrophic and atrophic signals linked to the
functional properties of the motor units that are likely to be imbalanced in HD. In this
article, we highlight the emerging field of research with particular focus on the recent
studies of the skeletal muscle pathology and the identification of new disease-modifying
treatments.
Keywords: Huntington’s disease, peripheral pathology, skeletal muscle atrophy, disease modifying treatment
INTRODUCTION
Huntington’s disease (HD) is neurodegenerative disorder caused
by the expansion of polyglutamine stretch within the huntingtin
protein (HTT) (Gusella et al., 1993; Vonsattel and DiFiglia, 1998;
Novak and Tabrizi, 2010). The disease is caused by the expan-
sion of a CAG repeat to over 35 CAG repeats in exon1 of the
huntingtin (HTT) gene which are normally observed in healthy
objects. Neurodegeneration, particularly widespread in the stri-
atal nuclei, basal ganglia and cereberal cortex, is a source of
neurological symptoms that involve motor, cognitive and psy-
chiatric disturbances (Novak and Tabrizi, 2010). This leads to
a wide-range of clinical features including personality changes,
motor impairment, dementia and weight loss that are likely to
progress over the course of 15–20 years to death (HDRG, 1993;
Walker, 2007). In mammals HTT is expressed in many tissues and
organs (Hoogeveen et al., 1993; Strong et al., 1993; Trottier et al.,
1995). HTT has been identified to be involved in many critical cel-
lular processes like transcription, protein trafficking and vesicle
transport (Li and Li, 2004). In mice HTT deletion is embryon-
ically lethal, leading to defects in all germ layers (Zeitlin et al.,
1995). It has been established that HTT is affecting organelles
and functional systems that are essential for all type cells i.e.,
mitochondria, ubuquitin-proteasome system and this phenom-
ena is not tissue specific (Li and Li, 2004; Sassone et al., 2009;
van der Burg et al., 2009; Zielonka et al., 2014). A recent study
of the heart function in two HD mouse models identified pro-
nounced contractile heart dysfunction, which might be a part of
dilatated cardiomyopathy (DCM). This was accompanied by the
re-expression of fetal genes, apoptotic cardiomyocyte loss and a
moderate degree of interstitial fibrosis (Mielcarek et al., 2014).
Therefore it is likely that the peripheral pathology of HD, such
as weight loss and severe skeletal muscle atrophy, might have a
significant input to the disease progression.
SKELETAL MUSCLE PATHOLOGY IN HD PATIENTS
A case-study report showed that a semi-professional marathon
runner (43 CAGs) developed signs of a slowly progressing myopa-
thy with elevated creatine kinase levels many years before first
signs of chorea were detected. Muscle biopsy revealed a mild
myopathy with mitochondrial pathology including a complex IV
deficiency (Kosinski et al., 2007). The isometric muscle strength
of 6 lower limb muscle groups was measured in 20 people with
HD and matched healthy controls. HD patients had reduced mus-
cle strength by 50% on average in comparison to healthy matched
controls (Busse et al., 2008). Several studies have reported defects
in the mitochondrial function of the central nervous system and
skeletal muscles in HD patients (Reddy, 2014). For example,
the non-invasive method of 31P-MRS (31P- Magnetic Resonance
Spectroscopy) showed a reduced phosphocreatine to inorganic
phosphate ratio in the symptomatic HD patients at rest. Muscle
www.frontiersin.org October 2014 | Volume 5 | Article 380 |1
e-mail: daniel.zielonka@gmail.com;
Zielonka et al. Atrophy of skeletal muscles in HD
ATP/phosphocreatine and inorganic phosphate levels were signif-
icantly reduced in both symptomatic and presymptomatic HD
subjects (Lodi et al., 2000). During recovery from exercise, the
maximum rate of mitochondrial ATP production was reduced
by 44% in the symptomatic HD patients and by 35% in the
presymptomatic HD carriers. HD subjects showed also a deficit
in the mitochondrial oxidative metabolism and that might sup-
port a role for mitochondrial dysfunction as a key factor involved
in the HD-related muscle pathogenesis (Lodi et al., 2000; Saft
et al., 2005). In addition, the total exercise capacity was normal
in HD subjects but notably the presymptomatic HD patients had
a lower anaerobic threshold and increased level of plasma lactate
(Ciammola et al., 2011). In vitro muscle cell cultures revealed that
HD cells produced more lactate and that might be indicative of
a higher glycolysis level (Ciammola et al., 2011). Furthermore,
muscle cultures showed cellular abnormalities including mito-
chondrial membrane potential, cytochrome c release, increased
CASPASE-3, 8, and 9 levels and defective cell differentiation,
likely due to the formation of HTT inclusions in differentiated
myotubes (Ciammola et al., 2006). Finally, electron microscopy
showed striking mitochondrial defects like abnormally elon-
gated and swollen mitochondria with derangement of cristae and
vacuoles (Ciammola et al., 2011).
ON THE WAY TO UNDERSTAND MUSCLE PATHOLOGY IN
HD—AN ANIMAL MODEL
Recent years of research on HD pathogenesis resulted in a gen-
eration of many HD transgenic mouse models including mHTT
N-terminal fragments and full-length murine or human mHTT
(Crook and Housman, 2011; Lee et al., 2013; Rattray et al., 2013).
R6/2 mice are transgenic for a mutated N-terminal exon 1 HTT
fragment and are the most frequently used in pre-clinical settings.
Behaviorally, R6/2 animals at first display a spatial learning deficit
at 3–4 weeks of age (Lione et al., 1999). Attention learning deficits
and abnormal performance in motor tests (swimming and high
speed rotarod) appear at 5–6 weeks of age (Carter et al., 1999;
Lione et al., 1999; Murphy et al., 2000), followed by development
of a resting tremor, gait disturbances and visual learning deficits
at 8–9 weeks of age (Carter et al., 1999; Murphy et al., 2000).
There is support that mHTT may have detrimental effects in the
skeletal muscles of R6/2 mice due to poly(Q) aggregate accumu-
lation (Sathasivam et al., 1999; Moffitt et al., 2009)andformation
of these inclusions in myoblasts and myotubes have been con-
firmed in vitro (Orth et al., 2003). Alternatively, the toxicity of
triplet repeat-containing RNA and/or patially mis-spliced hunt-
ingtin gene (Htt) could be considered as an additional mechanism
of HD pathology (Sathasivam et al., 2013).
Transcriptional deregulation is a typical feature of HD pathol-
ogy in the brain (Luthi-Carter et al., 2002). A similar transcrip-
tional profile in skeletal muscles (quadriceps) from R6/2 mice,
HdhQ150 homozygous knock-in mice and HD patients has been
identified and that was consistent with a transition from fast-
twitch to slow-twitch muscle fiber types (Luthi-Carter et al.,
2002). On the other hand, based on immunohistochemistry both
type I and II muscles were atrophic. Although atrophy occurred
in both type fibers, there was more type I fibers in the R6/2 skele-
tal muscles. Hence, there was a conversion of type II fibers to
type I during the process of muscle atrophy (Ribchester et al.,
2004). However, these findings in pre-clinical settings are incon-
sistent with an increased glycolysis observed in human patients
(Ciammola et al., 2011). Metabolic adaptations similar to those
induced by diabetes or fasting are also present in HD mouse
models but neither metabolic disorder could explain the full phe-
notype of HD muscle (Strand et al., 2005). Consequently, at
the ultrastructural level, the sciatic nerve displayed abnormali-
ties in large myelinated fibers in the presymptomatic R6/2 mice.
A significant decrease in the axoplasm diameter of myelinated
neurons and increased number of degenerating myelinated fibers
were observed; although myelin thickness and unmyelinated fiber
diameter were not affected (Wade et al., 2008). The synaptic
transmission at the neuromuscular junction has also been stud-
ied in the R6/1 mouse model of HD. The morphological data
suggest that the innervation pattern of the neuromuscular junc-
tions in R6/1 muscles were normal in early symptomatic animals.
However, the size and frequency of miniature endplate potentials
were not changed in the R6/1 mice, while the amplitude of evoked
endplate potentials increased. Consistent with a pre-synaptic
increase of release probability, synaptic depression under high-
frequency was higher in R6/1 mice. No changes were detected in
size and dynamics of the recycling synaptic vesicle pool (Rozas
et al., 2011). In contrast, it has been shown that skeletal muscles of
R6/2 mice developed age-dependent denervation-like abnormal-
ities, including reduced endplate area, supersensitivity to acetyl-
choline, decreased sensitivity to mu-conotoxin and anode-break
action potentials (Ribchester et al., 2004). Moreover, the minia-
ture endplate potential (mEPP) amplitude was notably increased
while mEPP frequency was significantly reduced in R6/2 mice.
Severely affected R6/2 mice developed a progressive increase
in a number of motor endplates that fail to respond to nerve
stimulation but there was no constitutive sprouting of motor
neurons, even in severely atrophic muscles. In fact there was
no age-dependent loss of regenerative capacity of motor neu-
rons in R6/2 mice (Ribchester et al., 2004). Another group has
studied the membrane properties of skeletal muscles that con-
trol contraction in the same HD mouse model. Adult skeletal
muscle from R6/2 mice showed that the action potentials in dis-
eased muscles were more easily triggered and prolonged than in
wild type littermates. Furthermore, the expression of the muscle
chloride channel (ClC-1) and Kcnj2 (Kir2.1 potassium chan-
nel) transcripts were significantly reduced and defects in mRNA
processing were detected (Waters et al., 2013).
To better understand a mechanism underlying muscle wast-
ing in the R6/2 mouse model, key pathways governing protein
metabolism, apoptosis and autophagy were examined. R6/2 mice
exhibited increased adiposity and elevated energy expenditure
without altered food intake. A total protein synthesis was unex-
pectedly increased in the gastrocnemius muscle by 19%, which
was associated with over-activation of rapamycin mTOR signal-
ing (She et al., 2011). The transcript levels of androgens, like
muscle ring finger-1 and atrophy F-box, were markedly attenu-
ated during fasting and re-feeding. Additionally, the mRNA level
of several caspase genes involved in both extrinsic and intrinsic
apoptotic pathways, like CASPASE-3/7, 8, and 9, were ele-
vated (She et al., 2011). Indeed, the CASPASE-6 up-regulation
Frontiers in Physiology | Striated Muscle Physiology October 2014 | Volume 5 | Article 380 |2
Zielonka et al. Atrophy of skeletal muscles in HD
might be due to enhanced activity of the p53 in the mus-
cles obtained from HD patients and from two different HD
mouse models. It has been also shown that CASPASE-6 may
target (cleave) laminin A (Ehrnhoefer et al., 2014). It was sug-
gested that this phenomenon might be mitigated by a small
molecule pifithrin-alpha, an inhibitor of p53 transcriptional
activity (Ehrnhoefer et al., 2014).
Since mitochondrial dysfunction might play a crucial role in
HD pathology (Quintanilla and Johnson, 2009),theroleofPPAR
γcoactivator 1α(PGC-1α) has been carefully assessed (Lin et al.,
2005). Reduced levels of PGC-1αand its target genes in skele-
tal muscles of HD transgenic mice and HD subjects have been
found. Treatment with guanidinopropionic acid (GPA) led to an
increased expression level of AMPK, PGC-1αtarget genes and
the genes characteristic for oxidative phosphorylation, electron
transport chain and mitochondrial biogenesis. Oxygen consump-
tion in response to GPA treatment was significantly reduced in
myoblasts from HD patients (Chaturvedi et al., 2009). On the
other hand, knockdown of mutant HTT resulted in increased
PGC-1αexpression in HD myoblast, while PGC-1αrescue led to
increased expression of markers for oxidative muscle fibers and
reversalofbluntedresponseforGPAinHDmice(Chaturvedi
et al., 2009). These findings showed that impaired function of
PGC-1αplays a critical role in the skeletal muscles dysfunction
in HD. Also, possible pharmacologic intervention with a small
molecule could enhance PGC-1αfunction may exert therapeu-
tic benefits (Chaturvedi et al., 2009). In addition, atrophic fibers
of R6/2 mice showed increased fuchsinophilic aggregates and
reduced cytochrome coxidase by 15%. Complex I–dependent
respiration of HD mitochondria showed more sensitivity to inhi-
bition by Ca2+than in wild-type mitochondria (Gizatullina et al.,
2006). A summary of morphological and molecular characteris-
tics of skeletal muscles in pre-clinical and clinical settings has been
presented in Table 1 .
CAN WE DELAY HD PROGRESSION BY MODULATING
MUSCLE FUNCTION?
As it has been mentioned in the previous paragraph, enhancing
PGC-1αactivity might be a good strategy to improve skeletal
muscles function in HD. Indeed, pharmacologic treatment with
the pan-PPAR agonist bezafibrate restored the PGC-1α,PPARs
Table 1 | Summary of defects observed in muscle in the pre-clinical
and clinical HD settings.
Human Mouse models
Reduced muscle strength √√
Muscle atrophy Unknown
Mitochondrial dysfunction √√
Inclusions formation √√
Transcriptional deregulation √√
Fast to slow twitch unknown
Increased of adiposity and protein synthesis unknown
Neuro-muscular junctions abnormalities unknown
Increased caspase activity √√
and downstream genes to wild type levels. It also prevented con-
versionoftypeIoxidativetotypeIIglycolyticmusclefibersas
well as increased muscle mitochondria numbers. Finally, bezafi-
brate rescued lipid accumulation and apparent vacuolization of
brown adipose tissue in the HD mice (Johri et al., 2012).
The other strategy to improve muscle function in HD is
based on the heat shock machinery modulation that could sup-
press mHTT aggregation (Labbadia and Morimoto, 2013). The
R6/2 mice expressing an active heat shock transcription factor
1 (HSF1) isoform had reduced polyglutamine inclusion forma-
tion and improved body weight. Unexpectedly, the lifespan of
R6/2:HSF1Tg mice were significantly improved despite the fact
that active HSF1 was not expressed in the brain. These results
indicated that active HSF1 has a strong inhibitory effect on polyg-
lutamine aggregates formation in vivo (Fujimoto et al., 2005).
Recent studies also showed that HDAC4 function in the
cytoplasm (Mielcarek et al., 2013a) and its reduction, delayed
cytoplasmic aggregate formation and rescued neuronal and
cortico-striatal synaptic function in HD mouse models. This was
accompanied by an improvement in motor co-ordination, neu-
rological phenotypes and increased lifespan (Mielcarek et al.,
2013b). Given that HDAC4 has well-established functions in
skeletal muscle, muscle atrophy is a major symptom of HD and
that HDAC4 has been linked to disease progression in an ALS
mouse model, it is likely that genetic reduction of HDAC4 in
skeletal muscle was a contributing factor to the improved HD
phenotypes (Bruneteau et al., 2013).
SUMMARY
HD is a complex disease that has a peripheral component to
its pathophysiology. Transcriptional changes in the HD skele-
tal muscles were comparable to those observed in the different
brain regions and skeletal muscle wasting/atrophy is likely to be
an important portion of HD pathogenesis. Some of the molec-
ular and physiological changes in HD muscles can be detected,
even in the pre-symptomatic HD individuals. On the molecu-
lar level, mitochondrial dysfunctions, PPAR alpha signaling and
HSF1 activation were identified as major players in the muscle
HD-related pathology. The major pathological pathways iden-
tiedinskeletalmuscleshavebeensummarizedinFigure 1.
However, many aspects of HD neuromuscular transmission and
FIGURE 1 | Summary of pathological events identified in the skeletal
muscles in HD.
www.frontiersin.org October 2014 | Volume 5 | Article 380 |3
Zielonka et al. Atrophy of skeletal muscles in HD
muscle physiology remain unanswered and need to be studied
more extensively. The proof of concept studies clearly showed that
by improving muscle function in HD mouse models, the progres-
sion of disease onset could be delayed and the lifespan extended.
Therefore this makes skeletal muscles an attractive target for
future therapies. Two key signaling pathways, i.e., Insulin like
growth Factor IGF and GDF-8/myostatin, have emerged in recent
years to be potent regulators of skeletal muscle size. Moreover,
several studies emphasized a role of hyperacetylation in muscle
wasting. Therefore there is a need for more pre-clinical and clini-
cal studies that will unravel the mechanism of HD skeletal muscles
pathology, leading to potential therapies in HD. Further work is
necessary in order to fully appreciate the complexity of the path-
ways that are affected during HD progression. Indeed, emerging
evidence has clearly indicated that peripheral tissues are as much
affected by the expression of the mutant huntingtin as the Central
Nervous System. Furthermore, the possibility to test the effect
of new drugs directly on human peripheral tissues is a new and
exciting research area.
REFERENCES
Bruneteau, G., Simonet, T., Bauche, S., Mandjee, N., Malfatti, E., Girard, E., et al.
(2013). Muscle histone deacetylase 4 upregulation in amyotrophic lateral scle-
rosis: potential role in reinnervation ability and disease progression. Brain 136,
2359–2368. doi: 10.1093/brain/awt164
Busse, M. E., Hughes, G., Wiles, C. M., and Rosser, A. E. (2008). Use of hand-
held dynamometry in the evaluation of lower limb muscle strength in people
with Huntington’s disease. J. Neurol. 255, 1534–1540. doi: 10.1007/s00415-008-
0964-x
Carter, R. J., Lione, L. A., Humby, T., Mangiarini, L., Mahal, A., Bates, G. P., et al.
(1999). Characterization of progressive motor deficits in mice transgenic for the
human Huntington’s disease mutation. J. Neurosci. 19, 3248–3257.
Chaturvedi, R. K., Adhihetty, P., Shukla, S., Hennessy, T., Calingasan, N., Yang, L.,
et al. (2009). Impaired PGC-1alpha function in muscle in Huntington’s disease.
Hum. Mol. Genet. 18, 3048–3065. doi: 10.1093/hmg/ddp243
Ciammola, A., Sassone, J., Alberti, L., Meola, G., Mancinelli, E., Russo, M. A., et al.
(2006). Increased apoptosis, Huntingtin inclusions and altered differentiation
in muscle cell cultures from Huntington’s disease subjects. Cell Death Differ. 13,
2068–2078. doi: 10.1038/sj.cdd.4401967
Ciammola, A., Sassone, J., Sciacco, M., Mencacci, N. E., Ripolone, M., Bizzi, C.,
et al. (2011). Low anaerobic threshold and increased skeletal muscle lactate pro-
duction in subjects with Huntington’s disease. Mov. Disord. 26, 130–137. doi:
10.1002/mds.23258
Crook, Z. R., and Housman, D. (2011). Huntington’s disease: can mice lead the way
to treatment? Neuron 69, 423–435. doi: 10.1016/j.neuron.2010.12.035
Ehrnhoefer, D. E., Skotte, N. H., Ladha, S., Nguyen, Y. T., Qiu, X., Deng, Y.,
et al. (2014). p53 increases caspase-6 expression and activation in muscle
tissue expressing mutant huntingtin. Hum. Mol. Genet. 23, 717–729. doi:
10.1093/hmg/ddt458
Fujimoto, M., Takaki, E., Hayashi, T., Kitaura, Y., Tanaka, Y., Inouye, S., et al.
(2005). Active HSF1 significantly suppresses polyglutamine aggregate forma-
tion in cellular and mouse models. J. Biol. Chem. 280, 34908–34916. doi:
10.1074/jbc.M506288200
Gizatullina, Z. Z., Lindenberg, K. S., Harjes, P., Chen, Y., Kosinski, C. M.,
Landwehrmeyer, B. G., et al. (2006). Low stability of Huntington muscle
mitochondria against Ca2+in R6/2 mice. Ann. Neurol. 59, 407–411. doi:
10.1002/ana.20754
Gusella, J. F., MacDonald, M. E., Ambrose, C. M., and Duyao, M. P. (1993).
Molecular genetics of Huntington’s disease. Arch. Neur ol. 50, 1157–1163. doi:
10.1001/archneur.1993.00540110037003
HDRG. (1993). A novel gene containing a trinucleotide repeat that is expanded
and unstable on Huntington’s disease chromosomes. The Huntington’s
Disease Collaborative Research Group. Cell 72, 971–83. doi: 10.1016/0092-
8674(93)90585-E
Hoogeveen, A. T., Willemsen, R., Meyer, N., de Rooij, K. E., Roos, R. A.,
van Ommen, G. J., et al. (1993). Characterization and localization of the
Huntington disease gene product. Hum. Mol. Genet. 2, 2069–2073. doi:
10.1093/hmg/2.12.2069
Johri, A., Calingasan, N. Y., Hennessey, T. M., Sharma, A., Yang, L., Wille, E.,
et al. (2012). Pharmacologic activation of mitochondrial biogenesis exerts
widespread beneficial effects in a transgenic mouse model of Huntington’s
disease. Hum. Mol. Genet. 21, 1124–1137. doi: 10.1093/hmg/ddr541
Kosinski, C. M., Schlangen, C., Gellerich, F. N., Gizatullina, Z., Deschauer, M.,
Schiefer, J., et al. (2007). Myopathy as a first symptom of Huntington’s disease
in a Marathon runner. Mov. Disord. 22, 1637–1640. doi: 10.1002/mds.21550
Labbadia, J., and Morimoto, R. I. (2013). Huntington’s disease: underlying molec-
ular mechanisms and emerging concepts. Trends Biochem. Sci. 38, 378–385. doi:
10.1016/j.tibs.2013.05.003
Lee, C. Y., Cantle, J. P., and Yang, X. W. (2013). Genetic manipulations of mutant
huntingtin in mice: new insights into Huntington’s disease pathogenesis. FEBS
J. 18, 4382–4394. doi: 10.1111/febs.12418
Li, S. H., and Li, X. J. (2004). Huntingtin-protein interactions and the
pathogenesis of Huntington’s disease. Tre n ds G e net . 20, 146–154. doi:
10.1016/j.tig.2004.01.008
Lin, J., Handschin, C., and Spiegelman, B. M. (2005). Metabolic control through
the PGC-1 family of transcription coactivators. Cell Metab. 1, 361–370. doi:
10.1016/j.cmet.2005.05.004
Lione, L. A., Carter, R. J., Hunt, M. J., Bates, G. P., Morton, A. J., and Dunnett, S.
B. (1999). Selective discrimination learning impairments in mice expressing the
human Huntington’s disease mutation. J. Neurosci. 19, 10428–10437.
Lodi, R., Schapira, A. H., Manners, D., Styles, P., Wood, N. W., Taylor, D. J., et al.
(2000). Abnormal in vivo skeletal muscle energy metabolism in Huntington’s
disease and dentatorubropallidoluysian atrophy. Ann. Neurol. 48, 72–76. doi:
10.1002/1531-8249(200007)48:1<72::AID-ANA11>3.0.CO;2-I
Luthi-Carter, R., Hanson, S. A., Strand, A. D., Bergstrom, D. A., Chun, W., Peters,
N. L., et al. (2002). Dysregulation of gene expression in the R6/2 model of polyg-
lutamine disease: parallel changes in muscle and brain. Hum. Mol. Genet. 11,
1911–1926. doi: 10.1093/hmg/11.17.1911
Mielcarek, M., Inuabasi, L., Bondulich, M. K., Muller, T., Osborne, G. F.,
Franklin, S. A., et al. (2014). Dysfunction of the CNS-Heart Axis in Mouse
Models of Huntington’s Disease. PLoS Genet. 10:e1004550. doi: 10.1371/jour-
nal.pgen.1004550
Mielcarek, M., Landles, C., Weiss, A., Bradaia, A., Seredenina, T., Inuabasi, L.,
et al. (2013b). HDAC4 reduction: a novel therapeutic strategy to target cyto-
plasmic huntingtin and ameliorate neurodegeneration. PLoS Biol. 11:e1001717.
doi: 10.1371/journal.pbio.1001717
Mielcarek, M., Seredenina, T., Stokes, M. P., Osborne, G. F., Landles, C., Inuabasi,
L., et al. (2013a). HDAC4 does not act as a protein deacetylase in the postnatal
murine brain in vivo.PLoS ONE 8:e80849. doi: 10.1371/journal.pone.0080849
Moffitt, H., McPhail, G. D., Woodman, B., Hobbs, C., and Bates, G. P. (2009).
Formation of polyglutamine inclusions in a wide range of non-CNS tissues
in the HdhQ150 knock-in mouse model of Huntington’s disease. PLoS ONE
4:e8025. doi: 10.1371/journal.pone.0008025
Murphy, K. P., Car ter, R. J., Lione, L. A., Mangiarini, L., Mahal, A., Bates, G. P., et al.
(2000). Abnormal synaptic plasticity and impaired spatial cognition in mice
transgenic for exon 1 of the human Huntington’s disease mutation. J. Neurosci.
20, 5115–5123.
Novak, M. J., and Tabrizi, S. J. (2010). Huntington’s disease. BMJ 340:c3109. doi:
10.1136/bmj.c3109
Orth, M., Cooper, J. M., Bates, G. P., and Schapira, A. H. (2003). Inclusion forma-
tion in Huntington’s disease R6/2 mouse muscle cultures. J. Neurochem. 87, 1–6.
doi: 10.1046/j.1471-4159.2003.02009.x
Quintanilla, R. A., and Johnson, G. V. W. (2009). Role of mitochondrial dysfunction
in the pathogenesis of Huntington’s disease. Brain Res. Bull. 80, 242–247. doi:
10.1016/j.brainresbull.2009.07.010
Rattray, I., Smith, E., Gale, R., Matsumoto, K., Bates, G. P., and Modo, M. (2013).
Correlations of behavioral deficits with brain pathology assessed through lon-
gitudinal MRI and histopathology in the R6/2 mouse model of HD. PLoS ONE
8:e60012. doi: 10.1371/journal.pone.0060012
Reddy, P. H. (2014). Increased mitochondrial fission and neuronal dys-
function in Huntington’s disease: implications for molecular inhibitors
of excessive mitochondrial fission. Drug Discov. Today 19, 951–955. doi:
10.1016/j.drudis.2014.03.020
Frontiers in Physiology | Striated Muscle Physiology October 2014 | Volume 5 | Article 380 |4
Zielonka et al. Atrophy of skeletal muscles in HD
Ribchester, R. R., Thomson, D., Wood, N. I., Hinks, T., Gillingwater, T. H., Wishart,
T. M., et al. (2004). Progressive abnormalities in skeletal muscle and neuromus-
cular junctions of transgenic mice expressing the Huntington’s disease muta-
tion. Eur. J. Neurosci. 20, 3092–3114. doi: 10.1111/j.1460-9568.2004.03783.x
Rozas, J. L., Gomez-Sanchez, L., Tomas-Zapico, C., Lucas, J. J., and Fernandez-
Chacon, R. (2011). Increased neurotransmitter release at the neuromuscular
junction in a mouse model of polyglutamine disease. J. neurosci. 31, 1106–1113.
doi: 10.1523/JNEUROSCI.2011-10.2011
Saft, C., Zange, J., Andrich, J., Muller, K., Lindenberg, K., Landwehrmeyer, B., et al.
(2005). Mitochondrial impairment in patients and asymptomatic mutation car-
riers of Huntington’s disease. Mov. Disord. 20, 674–679. doi: 10.1002/mds.20373
Sassone, J., Colciago, C., Cislaghi, G., Silani, V., and Ciammola, A. (2009).
Huntington’s disease: the current state of research with peripheral tissues. Exp.
Neuro l. 219, 385–397. doi: 10.1016/j.expneurol.2009.05.012
Sathasivam, K., Hobbs, C., Turmaine, M., Mangiarini, L., Mahal, A., Bertaux, F.,
et al. (1999). Formation of polyglutamine inclusions in non-CNS tissue. Hum.
Mol. Genet. 8, 813–822. doi: 10.1093/hmg/8.5.813
Sathasivam, K., Neueder,A., Gipson, T. A., Landles, C., Benjamin, A. C., Bondulich,
M. K., et al. (2013). Aberrant splicing of HTT generates the pathogenic exon 1
protein in Huntington disease. Proc. Natl. Acad. Sci. U.S.A. 110, 2366–2370. doi:
10.1073/pnas.1221891110
She, P., Zhang, Z., Marchionini, D., Diaz, W. C., Jetton, T. J., Kimball, S. R., et al.
(2011). Molecular characterization of skeletal muscle atrophy in the R6/2 mouse
model of Huntington’s disease. Am. J. Physiol. Endocrinol. Metab. 301, E49–E61.
doi: 10.1152/ajpendo.00630.2010
Strand, A. D., Aragaki, A. K., Shaw, D., Bird, T., Holton, J., Turner, C., et al. (2005).
Gene expression in Huntington’s disease skeletal muscle: a potential biomarker.
Hum. Mol. Genet. 14, 1863–1876. doi: 10.1093/hmg/ddi192
Strong, T. V., Tagle, D. A., Valdes, J. M., Elmer, L. W., Boehm, K., Swaroop, M., et al.
(1993). Widespread expression of the human and rat Huntington’s disease gene
in brain and nonneural tissues. Nat. Genet. 5, 259–265. doi: 10.1038/ng1193-259
Trottier, Y., Devys, D., Imbert, G., Saudou, F., An, I., Lutz, Y., et al. (1995). Cellular
localization of the Huntington’s disease protein and discrimination of the
normal and mutated form. Nat. Genet. 10, 104–110. doi: 10.1038/ng0595-104
van der Burg, J. M., Bjorkqvist, M., and Brundin, P. (2009). Beyond the brain:
widespread pathology in Huntington’s disease. Lancet Neurol. 8, 765–774. doi:
10.1016/S1474-4422(09)70178-4
Vonsattel, J. P., and DiFiglia, M. (1998). Huntington disease. J. Neuropathol. Exp.
Neuro l. 57, 369–384. doi: 10.1097/00005072-199805000-00001
Wade, A., Jacobs, P., and Morton, A. J. (2008). Atrophy and degeneration in sci-
atic nerve of presymptomatic mice carrying the Huntington’s disease mutation.
Brain Res. 1188, 61–68. doi: 10.1016/j.brainres.2007.06.059
Walker, F. O. (2007). Huntington’s disease. Lancet 369, 218–228. doi:
10.1016/S0140-6736(07)60111-1
Waters, C. W., Varuzhanyan, G., Talmadge, R. J., and Voss, A. A. (2013).
Huntington disease skeletal muscle is hyperexcitable owing to chloride and
potassium channel dysfunction. Proc. Natl. Acad. Sci. U.S.A. 110, 9160–9165.
doi: 10.1073/pnas.1220068110
Zeitlin, S., Liu, J. P., Chapman, D. L., Papaioannou, V. E., and Efstratiadis, A.
(1995). Increased apoptosis and early embryonic lethality in mice nullizygous
for the Huntington’s disease gene homologue. Nat. Genet. 11, 155–163. doi:
10.1038/ng1095-155
Zielonka, D., Piotrowska, I., and Mielcarek, M. (2014). Cardiac Dysfunction in
Huntington’s Disease. Exp. Clin. Cardiol. 20, 2547–2554. Available online at:
http://cardiologyacademicpress.com/soap/pdf/delme_1788_53e3f6e3313354.90
986595.pdf
Conflict of Interest Statement: Conflict of Interest Statement: The authors declare
that the research was conducted in the absence of any commercial or financial
relationships that could be construed as a potential conflict of interest.
Received: 31 July 2014; accepted: 13 September 2014; published online: 06 October
2014.
Citation: Zielonka D, Piotrowska I, Marcinkowski JT and Mielcarek M (2014) Skeletal
muscle pathology in Huntington’s disease. Front. Physiol. 5:380. doi: 10.3389/fphys.
2014.00380
This article was submitted to Striated Muscle Physiology, a section of the journal
Frontiers in Physiology.
Copyright © 2014 Zielonka, Piotrowska, Marcinkowski and Mielcarek. This is an
open-access article distributed under the terms of the Creative Commons Attribution
License (CC BY). The use, distribution or reproduction in other forums is permitted,
provided the original author(s) or licensor are credited and that the original publica-
tion in this journal is cited, in accordance with accepted academic practice. No use,
distribution or reproduction is permitted which does not comply with these terms.
www.frontiersin.org October 2014 | Volume 5 | Article 380 |5
... The results in Table 2 suggest alterations in circulating factors indicating abnormalities in skeletal muscle or generally energy metabolism in HD [74,75] or mouse models of HD [73]. Muscle metabolism is well known to be impaired in HD [4,30,78,79]. The deficits include mitochondrial dysfunction and reduced ATP production (assessed from phosphocreatine recovery), even in presymptomatic HD-mutation carriers [30,80]. ...
Article
Full-text available
Prominent pathological features of Huntington’s disease (HD) are aggregations of mutated Huntingtin protein (mHtt) in the brain and neurodegeneration, which causes characteristic motor (such as chorea and dystonia) and non-motor symptoms. However, the numerous systemic and peripheral deficits in HD have gained increasing attention recently, since those factors likely modulate disease progression, including brain pathology. While whole-body metabolic abnormalities and organ-specific pathologies in HD have been relatively well described, the potential mediators of compromised inter-organ communication in HD have been insufficiently characterized. Therefore, we applied an exploratory literature search to identify such mediators. Unsurprisingly, dysregulation of inflammatory factors, circulating mHtt, and many other messenger molecules (hormones, lipids, RNAs) were found that suggest impaired inter-organ communication, including of the gut–brain and muscle–brain axis. Based on these findings, we aimed to assess the risks and potentials of lifestyle interventions that are thought to improve communication across these axes: dietary strategies and exercise. We conclude that appropriate lifestyle interventions have great potential to reduce symptoms and potentially modify disease progression (possibly via improving inter-organ signaling) in HD. However, impaired systemic metabolism and peripheral symptoms warrant particular care in the design of dietary and exercise programs for people with HD.
... Huntington's disease (HD), a fatal neurodegenerative disorder characterized by progressive functional decline, is increasingly recognized for diverse pathology outside of the nervous system [1][2][3][4][5][6] . Mutant huntingtin protein is ubiquitously expressed throughout the body, an observation consistent with data suggesting that other tissues or cell types experience vulnerability and dysfunction. ...
Article
Full-text available
Huntington’s disease (HD) is increasingly recognized for diverse pathology outside of the nervous system. To describe the biology of HD in relation to functional progression, we previously analyzed the plasma and CSF metabolome in a cross-sectional study of participants who had various degrees of functional impairment. Here, we carried out an exploratory study in plasma from HD individuals over a 3-year time frame to assess whether differences exist between those with fast or absent clinical progression. There were more differences in circulating metabolite levels for fast progressors compared to absent progressors (111 vs 20, nominal p < 0.05). All metabolite changes in faster progressors were decreases, whereas some metabolite concentrations increased in absent progressors. Many of the metabolite levels that decreased in the fast progressors were higher at Screening compared to absent progressors but ended up lower by Year 3. Changes in faster progression suggest greater oxidative stress and inflammation (kynurenine, diacylglycerides, cysteine), disturbances in nitric oxide and urea metabolism (arginine, citrulline, ornithine, GABR), lower polyamines (putrescine and spermine), elevated glucose, and deficient AMPK signaling. Metabolomic differences between fast and absent progressors suggest the possibility of predicting functional decline in HD, and possibly delaying it with interventions to augment arginine, polyamines, and glucose regulation.
... This brain region is involved in motor control, emotion, habit formation, and reward [19]. Neuronal degeneration within the basal ganglia is consistent with the symptoms observed in HD patients, which include memory impairment, slurred speech, chorea, weight loss, and personality changes [20][21][22]. ...
Article
Full-text available
Huntington’s Disease (HD) is a severely debilitating neurodegenerative disorder in which sufferers exhibit different combinations of movement disorders, dementia, and behavioral or psychiatric abnormalities. The disorder is a result of a trinucleotide repeat expansion mutation that is inherited in an autosomal dominant manner. While there is currently no treatment to alter the course of HD, there are medications that lessen abnormal movement and psychiatric symptoms. ClinicalTrials.gov was searched to identify drugs that are currently in or have completed phase III drug trials for the treatment of HD. The described phase III trials were further limited to interventional studies that were recruiting, active not recruiting, or completed. In addition, all studies must have posted an update within the past year. PubMed was used to gather further information on these interventional studies. Of the nine clinical trials that met these criteria, eight involved the following drugs: metformin, dextromethorphan/quinidine, deutetrabenazine, valbenazine, Cellavita HD, pridopidine, SAGE-718, and RO7234292 (RG6042). Of these drug treatments, four are already FDA approved. This systematic review provides a resource that summarizes the present therapies for treating this devastating condition that are currently in phase III clinical trials in the United States.
... The importance of huntingtin in the periphery is supported by the finding that a mouse model expressing mutant huntingtin ubiquitously has severe cardiac dysfunction (Mihm et al., 2007). Beyond the heart, there is significant evidence that HTT is important in skeletal muscle, as movement dysfunction and skeletal muscle weakness are hallmarks of HD (Busse et al., 2008;Zielonka et al., 2014). Additionally, it is possible that brain-muscle crosstalk contributes to dysfunction in HD (Chuang and Demontis, 2021), especially with recent evidence showing that muscle-specific manipulations can affect brain function (Ehlen et al., 2017). ...
Article
Full-text available
Huntington’s disease (HD) is a neurodegenerative disease characterized by movement and cognitive dysfunction. HD is caused by a CAG expansion in exon 1 of the HTT gene that leads to a polyglutamine (PQ) repeat in the huntingtin protein, which aggregates in the brain and periphery. Previously, we used Drosophila models to determine that Htt-PQ aggregation in the heart causes shortened lifespan and cardiac dysfunction that is ameliorated by promoting chaperonin function or reducing oxidative stress. Here, we further study the role of neuronal mutant huntingtin and how it affects peripheral function. We overexpressed normal ( Htt-PQ25 ) or expanded mutant ( Htt-PQ72 ) exon 1 of huntingtin in Drosophila neurons and found that mutant huntingtin caused age-dependent Htt-PQ aggregation in the brain and could cause a loss of synapsin. To determine if this neuronal dysfunction led to peripheral dysfunction, we performed a negative geotaxis assay to measure locomotor performance and found that neuronal mutant huntingtin caused an age-dependent decrease in locomotor performance. Next, we found that rapamycin reduced Htt-PQ aggregation in the brain. These results demonstrate the role of neuronal Htt-PQ in dysfunction in models of HD, suggest that brain-periphery crosstalk could be important to the pathogenesis of HD, and show that rapamycin reduces mutant huntingtin aggregation in the brain.
... Since HD patients often present bioenergetic deficits and pathology of skeletal muscles (Lodi et al., 2000;Zielonka et al., 2014;Djoussé et al., 2002;Kosinski et al., 2007;Saft et al., 2005), we also measured mitochondrial respiration in two-week old flies expressing HTT93Q in the muscles via the Mef2GAL4 driver (Mef2 > HTT93Q). These flies exhibited highly elevated complex I-linked LEAK. ...
Article
Huntington's disease (HD) is a fatal neurodegenerative disease with limited treatment options. Human and animal studies have suggested that metabolic and mitochondrial dysfunctions contribute to HD pathogenesis. Here, we use high-resolution respirometry to uncover defective mitochondrial oxidative phosphorylation and electron transfer capacity when a mutant huntingtin fragment is targeted to neurons or muscles in Drosophila and find that enhancing mitochondrial function can ameliorate these defects. In particular, we find that co-expression of parkin, an E3 ubiquitin ligase critical for mitochondrial dynamics and homeostasis, produces significant enhancement of mitochondrial respiration when expressed either in neurons or muscles, resulting in significant rescue of neurodegeneration, viability and longevity in HD model flies. Targeting mutant HTT to muscles results in larger mitochondria and higher mitochondrial mass, while co-expression of parkin increases mitochondrial fission and decreases mass. Furthermore, directly addressing HD-mediated defects in the fly's mitochondrial electron transport system, by rerouting electrons to either bypass mitochondrial complex I or complexes III-IV, significantly increases mitochondrial respiration and results in a striking rescue of all phenotypes arising from neuronal mutant huntingtin expression. These observations suggest that bypassing impaired mitochondrial respiratory complexes in HD may have therapeutic potential for the treatment of this devastating disorder.
... Continued deterioration of the striatal component of the basal ganglia and cortical neurons manifests as motor, cognitive, behavioral, and affective symptoms that ultimately lead to death within 20 years of symptom onset (5). Interestingly, though skeletal muscle wasting is a hallmark of Huntington's disease, the mechanism underlying this atrophy is unknown, but is postulated to be caused by disrupted cell-cell signaling due to abnormal production of the Huntingtin protein (6). Motor manifestations of the disease, such as muscle rigidity and involuntary jerking movements, may contribute to muscle breakdown. ...
Article
Full-text available
Background Rhabdomyolysis-induced acute kidney injury is a serious condition that can progress to acute renal failure if not promptly identified and treated. Rhabdomyolysis occurs when serum creatine kinase levels approach > 1000 U/L (five times the normal upper limit). The chance of acute kidney injury increases as the levels of creatine kinase increase. Although Huntington’s disease is associated with muscle atrophy, elevated baseline creatine kinase levels in these patients have not been routinely reported. Case presentation A 31-year-old African American patient presented to the emergency department after he was found unconscious from a fall attributed to the progression of his Huntington’s disease. On admission, he had an extremely high creatine kinase level of 114,400 U/L and was treated with fluids, electrolyte balance, and dialysis. However, his condition progressed to acute renal failure and he later developed posterior reversible encephalopathy syndrome, requiring transfer to the intensive care unit with placement on continuous renal replacement therapy. Eventually, his kidney function recovered and he was discharged home with 24/7 care by his family for persistent impairments related to his Huntington’s disease. Conclusions This case report underscores the importance of promptly recognizing elevated creatine kinase levels in patients with Huntington’s disease due to the risk of developing rhabdomyolysis-induced acute kidney injury. If not aggressively treated, the condition of these patients is likely to progress to renal failure. Anticipating the progression of rhabdomyolysis-induced acute kidney injury is paramount to improving clinical outcomes. Additionally, this case identifies a potential link between the patient’s Huntington’s disease and his abnormally elevated creatine kinase, a finding not described in the literature of rhabdomyolysis-induced kidney injuries to date and an important consideration for future patients with similar comorbidities.
Article
Full-text available
Neuron-to-neuron transmission of aggregation-prone, misfolded proteins may potentially explain the spatiotemporal accumulation of pathological lesions in the brains of patients with neurodegenerative protein-misfolding diseases (PMDs). However, little is known about protein transmission from the central nervous system to the periphery, or how this propagation contributes to PMD pathology. To deepen our understanding of these processes, we established two functional neuromuscular systems derived from human iPSCs. One was suitable for long-term high-throughput live-cell imaging and the other was adapted to a microfluidic system assuring that connectivity between motor neurons and muscle cells was restricted to the neuromuscular junction. We show that the Huntington's disease (HD)-associated mutant HTT exon 1 protein (mHTTEx1) is transmitted from neurons to muscle cells across the human neuromuscular junction. We found that transmission is an active and dynamic process that starts before aggregate formation and is regulated by synaptic activity. We further found that transmitted mHTTEx1 causes HD-relevant pathology at both molecular and functional levels in human muscle cells, even in the presence of the ubiquitous expression of mHTTEx1. In conclusion, we have uncovered a causal link between mHTTEx1 synaptic transmission and HD pathology, highlighting the therapeutic potential of blocking toxic protein transmission in PMDs.
Chapter
A disease is a multifactorial dysfunction of system that affects the structure and function in an organism. In the same way, neurological disorders are associated with multiple factors like gene overexpression, repression, complete knockout, protein misfolding, metabolic dysfunction, microbial infection, or dysbiosis of microbiome. It is an established fact that several neurodegenerative, neurobehavioural, mental, and metabolic disorders including Alzheimer’s, Parkinson’s, Huntington’s, schizophrenia, multiple sclerosis, depression, and obesity have been allied to microbiota. Microbiome solely might not be the cause of any disease, but its contribution is significant. The molecular interactions between gut microbiome and nervous system are complex and bidirectional; any alterations in the gut–brain axis might leads to gastrointestinal and neurological disorders. It is easy to manipulate gut microbiome rather than the brain for better therapies, so this chapter deals with microbiome association in development of neurological disorders.KeywordsGutMicrobiomeNeurodegenerationDisordersMultiple sclerosisAlzheimer’s
Article
Full-text available
Huntington's disease (HD) is a multi-tissue failure disorder for which there is no cure. We have previously shown an effective therapeutic approach limited mainly to the central nervous system, based on a synthetic zinc finger (ZF) transcription repressor gene therapy, but it would be important to target other tissues as well. In this study, we identify a novel minimal HSP90AB1 promoter region that can efficiently control expression not only in the CNS but also in other affected HD tissues. This promoter-enhancer is effective in driving expression of ZF therapeutic molecules in both HD skeletal muscles and the heart, in the symptomatic R6/1 mouse model. Moreover, for the first time we show that ZF molecules repressing mutant HTT reverse transcriptional pathological remodelling in HD hearts. We conclude that this HSP90AB1 minimal promoter may be used to target multiple HD organs with therapeutic genes. The new promoter has the potential to be added to the portfolio of gene therapy promoters, for use where ubiquitous expression is needed.
Article
Full-text available
Huntington's disease (HD) is a progressive neurological disorder for which there is no effective disease?modifying treatment (HTT). HTT is ubiquitiniusly expressed in many tissues and therefore might be also associated with abnormalities in the peripheral organs. It is not fully understood whether these pathological changes are primary to the brain dysfunction or are secondary to the central nervous system malfunctions. It has become more apparent that HD patients experience a wide array of peripheral organs dysfunction and HD patients exhibit a high incidence of cardiovascular events, presumably as a result of accumulation of toxic amyloid?like inclusions. In this article, we highlight this emerging field of research with particular focus on the recent studies on the cardiac pathology.
Article
Full-text available
Cardiac remodelling and contractile dysfunction occur during both acute and chronic disease processes including the accumulation of insoluble aggregates of misfolded amyloid proteins that are typical features of Alzheimer's, Parkinson's and Huntington's disease (HD). While HD has been described mainly as a neurological disease, multiple epidemiological studies have shown that HD patients exhibit a high incidence of cardiovascular events leading to heart failure, and that this is the second highest cause of death. Given that huntingtin is ubiquitously expressed, cardiomyocytes may be at risk of an HD-related dysfunction. In mice, the forced expression of an expanded polyQ repeat under the control of a cardiac specific promoter led to severe heart failure followed by reduced lifespan. However the mechanism leading to cardiac dysfunction in the clinical and pre-clinical HD settings remains unknown. To unravel this mechanism, we employed the R6/2 transgenic and HdhQ150 knock-in mouse models of HD. We found that pre-symptomatic animals developed connexin-43 relocation and a significant deregulation of hypertrophic markers and Bdnf transcripts. In the symptomatic animals, pronounced functional changes were visualised by cardiac MRI revealing a contractile dysfunction, which might be a part of dilatated cardiomyopathy (DCM). This was accompanied by the re-expression of foetal genes, apoptotic cardiomyocyte loss and a moderate degree of interstitial fibrosis. To our surprise, we could identify neither mutant HTT aggregates in cardiac tissue nor a HD-specific transcriptional dysregulation, even at the end stage of disease. We postulate that the HD-related cardiomyopathy is caused by altered central autonomic pathways although the pathogenic effects of mutant HTT acting intrinsically in the heart may also be a contributing factor.
Data
Full-text available
Huntington's disease (HD) is caused by the expansion of a CAG repeat in the huntingtin (HTT) gene. The R6 mouse models of HD express a mutant version of exon 1 HTT and typically develop motor and cognitive impairments, a widespread huntingtin (HTT) aggregate pathology and brain atrophy. Unlike the more commonly used R6/2 mouse line, R6/1 mice have fewer CAG repeats and, subsequently, a less rapid pathological decline. Compared to the R6/2 line, fewer descriptions of the progressive pathologies exhibited by R6/1 mice exist. The association between the molecular and cellular neuropathology with brain atrophy, and with the development of behavioral phenotypes remains poorly understood in many models of HD. In attempt to link these factors in the R6/1 mouse line, we have performed detailed assessments of behavior and of regional brain abnormalities determined through longitudinal, in vivo magnetic resonance imaging (MRI), as well as an end-stage, ex vivo MRI study and histological assessment. We found progressive decline in both motor and non-motor related behavioral tasks in R6/1 mice, first evident at 11 weeks of age. Regional brain volumes were generally unaffected at 9 weeks, but by 17 weeks there was significant grey matter atrophy. This age-related brain volume loss was validated using a more precise, semi-automated Tensor Based morphometry assessment. As well as these clear progressive phenotypes, mutant HTT (mHTT) protein, the hallmark of HD molecular pathology, was widely distributed throughout the R6/1 brain and was accompanied by neuronal loss. Despite these seemingly concomitant, robust pathological phenotypes, there appeared to be little correlation between the three main outcome measures: behavioral performance, MRI-detected brain atrophy and histopathology. In conclusion, R6/1 mice exhibit many features of HD, but the underlying mechanisms driving these clear behavioral disturbances and the brain volume loss, still remain unclear.
Article
Full-text available
Author Summary Huntington's disease (HD) is a late-onset neurodegenerative disorder caused by protein-folding defects in the huntingtin protein. Mutations in huntingtin can result in extra-long tracts of the amino acid glutamine, resulting in aberrant interactions with other proteins and also causing huntingtin proteins to self-associate and -aggregate. The pathology of HD is therefore associated with nuclear and cytoplasmic aggregates. HDAC4 is a histone deacetylase protein traditionally associated with roles in transcription repression. The HDAC4 protein contains a glutamine-rich domain and in this work we find that HDAC4 associates with huntingtin in a polyglutamine-length-dependent manner and that these proteins co-localise in cytoplasmic inclusions. Importantly, reducing HDAC4 levels delays cytoplasmic aggregate formation and rescues neuronal and cortico-striatal synaptic function in mouse models of HD. In addition, we observe improvements in motor coordination and neurological phenotypes, as well as increased lifespan in these mice. Nuclear huntingin aggregates or transcription regulation, however, remained unaffected when HDAC4 levels were reduced to enable these effects. Our results thus provide valuable insight into separating cytoplasmic and nuclear pathologies, and define a crucial role for cytoplasmic aggregations in HD progression. HDAC4 reduction presents a novel strategy for alleviating the toxicity of huntingtin protein aggregation, thereby influencing the molecular pathology of Huntington's disease. As there are currently no disease-modifying therapeutics available for Huntington's disease, we hope that this HDAC4-mediated regulation may be amenable to small-molecule therapeutics.
Article
Full-text available
Reversible protein acetylation provides a central mechanism for controlling gene expression and cellular signaling events. It is governed by the antagonistic commitment of two enzymes families: the histone acetyltransferases (HATs) and the histone deacetylases (HDACs). HDAC4, like its class IIa counterparts, is a potent transcriptional repressor through interactions with tissue specific transcription factors via its N-terminal domain. Whilst the lysine deacetylase activity of the class IIa HDACs is much less potent than that of the class I enzymes, HDAC4 has been reported to influence protein deacetylation through its interaction with HDAC3. To investigate the influence of HDAC4 on protein acetylation we employed the immunoaffinity-based AcetylScan proteomic method. We identified many proteins known to be modified by acetylation, but found that the absence of HDAC4 had no effect on the acetylation profile of the murine neonate brain. This is consistent with the biochemical data suggesting that HDAC4 may not function as a lysine deacetylase, but these in vivo data do not support the previous report showing that the enzymatic activity of HDAC3 might be modified by its interaction with HDAC4. To complement this work, we used Affymetrix arrays to investigate the effect of HDAC4 knock-out on the transcriptional profile of the postnatal murine brain. There was no effect on global transcription, consistent with the absence of a differential histone acetylation profile. Validation of the array data by Taq-man qPCR indicated that only protamine 1 and Igfbp6 mRNA levels were increased by more than one-fold and only Calml4 was decreased. The lack of a major effect on the transcriptional profile is consistent with the cytoplasmic location of HDAC4 in the P3 murine brain.
Article
Full-text available
Activation of caspase-6 in the striatum of both presymptomatic and affected persons with Huntington's disease (HD) is an early event in the disease pathogenesis. However, little is known about the role of caspase-6 outside the central nervous system (CNS) and whether caspase activation might play a role in the peripheral phenotypes, such as muscle wasting observed in HD. We assessed skeletal muscle tissue from HD patients and well-characterized mouse models of HD. Cleavage of the caspase-6 specific substrate lamin A is significantly increased in skeletal muscle obtained from HD patients as well as in muscle tissues from two different HD mouse models. p53, a transcriptional activator of caspase-6, is upregulated in neuronal cells and tissues expressing mutant huntingtin. Activation of p53 leads to a dramatic increase in levels of caspase-6 mRNA, caspase-6 activity and cleavage of lamin A. Using mouse embryonic fibroblasts (MEFs) from YAC128 mice, we show that this increase in caspase-6 activity can be mitigated by pifithrin-α (pifα), an inhibitor of p53 transcriptional activity, but not through the inhibition of p53's mitochondrial pro-apoptotic function. Remarkably, the p53-mediated increase in caspase-6 expression and activation is exacerbated in cells and tissues of both neuronal and peripheral origin expressing mutant huntingtin (Htt). These findings suggest that the presence of the mutant Htt protein enhances p53 activity and lowers the apoptotic threshold, which activates caspase-6. Furthermore, these results suggest that this pathway is activated both within and outside the CNS in HD and may contribute to both loss of CNS neurons and muscle atrophy.
Article
Full-text available
Amyotrophic lateral sclerosis is a typically rapidly progressive neurodegenerative disorder affecting motor neurons leading to progressive muscle paralysis and death, usually from respiratory failure, in 3-5 years. Some patients have slow disease progression and prolonged survival, but the underlying mechanisms remain poorly understood. Riluzole, the only approved treatment, only modestly prolongs survival and has no effect on muscle function. In the early phase of the disease, motor neuron loss is initially compensated for by collateral reinnervation, but over time this compensation fails, leading to progressive muscle wasting. The crucial role of muscle histone deacetylase 4 and its regulator microRNA-206 in compensatory reinnervation and disease progression was recently suggested in a mouse model of amyotrophic lateral sclerosis (transgenic mice carrying human mutations in the superoxide dismutase gene). Here, we sought to investigate whether the microRNA-206-histone deacetylase 4 pathway plays a role in muscle compensatory reinnervation in patients with amyotrophic lateral sclerosis and thus contributes to disease outcome differences. We studied muscle reinnervation using high-resolution confocal imaging of neuromuscular junctions in muscle samples obtained from 11 patients with amyotrophic lateral sclerosis, including five long-term survivors. We showed that the proportion of reinnervated neuromuscular junctions was significantly higher in long-term survivors than in patients with rapidly progressive disease. We analysed the expression of muscle candidate genes involved in the reinnervation process and showed that histone deacetylase 4 upregulation was significantly greater in patients with rapidly progressive disease and was negatively correlated with the extent of muscle reinnervation and functional outcome. Conversely, the proposed regulator of histone deacetylase 4, microRNA-206, was upregulated in both patient groups, but did not correlate with disease progression or reinnervation. We conclude that muscle expression of histone deacetylase 4 may be a key factor for muscle reinnervation and disease progression in patients with amyotrophic lateral sclerosis. Specific histone deacetylase 4 inhibitors may then constitute a therapeutic approach to enhancing motor performance and slowing disease progression in amyotrophic lateral sclerosis.
Article
Huntington's disease (HD) is a fatal, progressive neurodegenerative disease with an autosomal dominant inheritance, characterized by chorea, involuntary movements of the limbs and cognitive impairments. Since identification of the HD gene in 1993, tremendous progress has been made in identifying underlying mechanisms involved in HD pathogenesis and progression, and in developing and testing molecular therapeutic targets, using cell and animal models of HD. Recent studies have found that mutant Huntingtin (mHtt) interacts with Dynamin-related protein 1 (Drp1), causing excessive fragmentation of mitochondria, leading to abnormal mitochondrial dynamics and neuronal damage in HD-affected neurons. Some progress has been made in developing molecules that can reduce excessive mitochondrial fission while maintaining both the normal balance between mitochondrial fusion and fission, and normal mitochondrial function in diseases in which excessive mitochondrial fission has been implicated. In this article, we highlight investigations that are determining the involvement of excessive mitochondrial fission in HD pathogenesis, and that are developing inhibitors of excessive mitochondrial fission for potential therapeutic applications.
Article
This year (2013) marks the twentieth anniversary of the identification of the causal genetic mutation for Huntington's disease (HD), a landmark discovery that has heralded the study of the biological underpinnings of this most common dominantly inherited neurodegenerative disorder. Among the variety of model organisms used to study HD pathogenesis, the mouse model has been by far the most commonly used mammalian genetic model organism. Much of our current knowledge regarding mutant Huntingtin (mHtt)-induced disease pathogenesis in mammalian models has been gained from studying transgenic mouse models expressing mHtt N-terminal fragments, full-length murine or human mHtt. In this review, we will focus on recent progress in using novel HD mouse models with targeted mHtt expression in specific brain cell types. These models help to address the role of distinct neuronal and non-neuronal cell types in eliciting cell-autonomous or non-cell-autonomous disease processes in HD. We will also describe several mHtt transgenic mouse models with targeted mutations in Htt cis-domains to address specific pathogenic hypotheses, ranging from mHtt proteolysis to post-translational modifications. These novel mouse genetic studies, through direct manipulations of the causal HD gene, provide a reductionist approach to systematically unravel the cellular and molecular pathways that are targeted by mHtt in disease pathogenesis and to potentially identify novel targets for therapy. This article is protected by copyright. All rights reserved.
Article
Huntington's disease (HD) is a progressive neurodegenerative disorder for which no disease modifying treatments exist. Many molecular changes and cellular consequences that underlie HD are observed in other neurological disorders, suggesting that common pathological mechanisms and pathways may exist. Recent findings have enhanced our understanding of the way cells regulate and respond to expanded polyglutamine proteins such as mutant huntingtin. These studies demonstrate that in addition to effects on folding, aggregation, and clearance pathways, a general transcriptional mechanism also dictates the expression of polyglutamine proteins. Here, we summarize the key pathways and networks that are important in HD in the context of recent therapeutic advances and highlight how their interplay may be of relevance to other protein folding disorders.