ArticlePDF Available

Cognitive Function in Health and Disease: The Role of Epigenetic Mechanisms

Authors:
  • Eisai AiM Institute, Eisai Inc.

Abstract and Figures

Epigenetic mechanisms regulate the interaction between the genome and the environment and have been implicated in the etiology of various brain diseases. One type of epigenetic modification, histone acetylation, is dynamically altered during memory formation. Histone acetylation is regulated by the activities of histone deacetylase (HDAC) and histone acetyltransferase enzymes. The use of HDAC inhibitors has emerged as a promising new strategy for the therapeutic intervention of neurodegenerative disease. We used a combination of pharmacological and mouse genetic approaches that allowed us to identify HDAC2 as a specific negative regulator of synaptic plasticity and memory formation. Our results suggest that HDAC inhibitors enhance cognitive function by inhibiting HDAC2, which renders HDAC2 target genes more accessible to transcriptional activators and coactivators recruited by neuronal activity stimulation. The data presented at the 2011 Barcelona ADPD Conference delineate a novel and important role for HDAC2 activity in the cognitive impairments associated with neurodegenerative disease.
Content may be subject to copyright.
Fax +41 61 306 12 34
E-Mail karger@karger.ch
www.karger.com
Neurodegenerative Dis
DOI: 10.1159/000334602
Cognitive Function in Health and Disease:
The Role of Epigenetic Mechanisms
Alison E. Mungenast a Li-Huei Tsai a–c
a Picower Institute for Learning and Memory,
b Department of Brain and Cognitive Sciences, Massachusetts Institute
of Technology, and
c Howard Hughes Medical Institute, Cambridge, Mass. , USA
The Epigenetic Regulation of Gene Transcription
Epigenetic mechanisms, such as histone modification
and DNA methylation, are key regulators of gene-envi-
ronment interactions and have been implicated in the eti-
ology of various brain diseases [for a review, see ref.
1 ].
One common form of epigenetic modification is histone
acetylation, which is dynamically altered during memory
formation
[2] . Long-lasting forms of synaptic plasticity
involve changes in the expression of genes involved in
synaptic functioning [for a review, see ref.
3 ] and, in re-
cent years, accumulating evidence indicates that epige-
netic mechanisms play crucial roles in promoting these
long-lasting changes
[4] .
Histone proteins may become acetylated on lysine res-
idues. The addition of an acetyl group leads to a relax-
ation of the chromatin that has been associated with tran-
scriptional activation
[5] . Histone acetylation is regulated
by the opposing activities of histone deacetylase (HDAC)
and histone acetyltransferase enzymes
[6, 7] . Class I
HDACs are primarily found within the nucleus, where
they regulate histone acetylation and suppress gene ex-
pression
[8] . Alterations in histone acetylation have re-
cently been shown to have an important role during
learning and memory processes. Numerous laboratories
have shown that pharmacological HDAC inhibitors
Key Words
Alzheimer’s disease Synaptic plasticity Learning and
memory Epigenetics Chromatin Histone deacetylase
Abstract
Epigenetic mechanisms regulate the interaction between
the genome and the environment and have been implicated
in the etiology of various brain diseases. One type of epige-
netic modification, histone acetylation, is dynamically al-
tered during memory formation. Histone acetylation is regu-
lated by the activities of histone deacetylase (HDAC) and his-
tone acetyltransferase enzymes. The use of HDAC inhibitors
has emerged as a promising new strategy for the therapeutic
intervention of neurodegenerative disease. We used a com-
bination of pharmacological and mouse genetic approaches
that allowed us to identify HDAC2 as a specific negative reg-
ulator of synaptic plasticity and memory formation. Our re-
sults suggest that HDAC inhibitors enhance cognitive func-
t io n b y in hi b it i ng HD AC 2 , w hi c h r e nd er s HD AC 2 ta rg e t g e ne s
more accessible to transcriptional activators and coactiva-
tors recruited by neuronal act ivit y stimu lation . The data pre-
sented at the 2011 Barcelona ADPD Conference delineate a
novel and important role for HDAC2 activity in the cognitive
impairments associated with neurodegenerative disease.
Copyr ight © 2012 S. Karger AG, Ba sel
Recei ved: July 1, 2011
Accepted a fter revision: November 25, 2011
Publish ed online: February 1, 2012
Diseases
Li-Huei Tsai
Howard Hughes Medical Ins titute
77 Massachusetts Avenue
Cambridge, M A 02139 (USA)
E-Mail lhtsai @ mit.edu
© 2012 S. Ka rger AG, Basel
1660–2854/12/0000–0000$38.00/0
Accessible online at:
www.karger.com/ndd
© Free Author
Copy – for per-
sonal use only
ANY DISTRIBUTION OF THIS
ARTICLE WITHOUT WRITTEN
CONSENT FROM S. KARGER
AG, BASEL IS A VIOLATION
OF THE COPYRIGHT.
Written permission to distrib-
ute the PDF will be granted
against payment of a per-
mission fee, which is based
on the number of accesses
required. Please contact
permission@karger.ch
© Free Author Copy – for per sonal use only
ANY DISTRIBUTION OF THIS ARTICLE WITHOUT WRITTEN CONSENT FROM S. KARGER AG, BASEL IS A VIOLATION OF THE COPYRIGHT.
Written permission to distribute the PDF will be granted against payment of a per mission fee, which is based on the number of accesses required. Please contact permission@karger.ch
Mungenast /Tsai
Neurodegenerative Dis
2
(HDACis) improve cognition in both wild-type animals
and in animal models of neurological disease, and that
HDACis targeting class I HDACs (HDACs 1, 2, 3 and 8),
such as suberoylanilide hydroxamic acid (SAHA), are the
most effective in enhancing cognitive function [for a re-
view, see ref.
9 ]. Guan et al. [4] used SAHA-based affinity
probes
[10] to identify the closely related HDAC1 and
HDAC2 as the main cellular targets of SAHA, followed
by a genetic approach to dissect out the contribution
of HDAC2 versus HDAC1 to cognitive function. These
studies show that transgenic mice that have a neuron-
specific overexpression of HDAC2 exhibit striking im-
pairments in memory formation and synaptic plasticity
that ar e not obser ved i n HDAC1-overex press ing mic e
[4] .
HDAC2 knockout (KO) mice, but not HDAC1 KO mice,
exhibit enhanced synaptic plasticity and facilitated learn-
ing and memory, and serve as a rare genetic model of
cognitive enhancement. These findings indicate that
HDAC2 plays a major role in learning and memory and
synaptic plasticity, as well as in the acetylation of the age-
associated H4K12 residue.
p25/Cdk5 and Neurodegeneration
The proline-directed serine/threonine kinase, Cdk5,
has been well characterized as a tau kinase
[11, 12] that
associates with neurofibrillary tangles in the brain of
Alzheimer’s disease (AD) patients
[13 , 14] . The activation
of Cdk5 results from its association with one of two acti-
vators, p35 or p39
[15 , 16] . The truncated form of p35, p25,
was shown to be increased in the brains of AD patients
[17,
18] , and the increased generation of p25 was subsequently
found in mouse models of AD [for a review, see ref.
19 ],
ischemia
[20] , amyotrophic lateral sclerosis [21] , Parkin-
son’s disease [22] , Huntington’s disease [23] , and hippo-
campal sclerosis
[24] . The cleavage of p35 to p25 is induced
in primary neurons upon exposure to a number of neuro-
toxic insults, which include oxidative stress (H
2 O 2 ), exci-
totoxicity ( glutamate), high interna l calcium (ionophores),
and -amyloid protein
[20] . Therefore, p25 generation is
associated with neurotoxicity both in vitro and in vivo.
The bi-transgenic CK-p25 mouse model of neurode-
generation expresses a p25- green fluorescent protein fu-
sion protein in an inducible, postnatal and forebrain-spe-
cific manner
[25] . Upon the induction of p25 expression,
the sequelae of neurodegeneration occur in a rapid and
predictable manner
[25, 26] . In the CK-p25 mouse, p25
overexpression is associated with the eventual death of
nearly 40% of hippocampal and cortical neurons.
HDACi Treatment Reinstates Learning and Restores
Remote Memory after Neurodegeneration
The treatment of 6-week-induced CK-p25 mice, which
display severe neurodegeneration, with the non-selective
HDACi sodium butyrate significantly improves cogni-
tive function
[27] . The HDACis SAHA and phenylbuty-
rate also have the capacity to reinstate learning behavior
in the APPswe/PS1dE9 mouse model of AD
[28, 29] .
HDAC inhibition, which resulted in the recovery of learn-
ing ability, did not affect amyloid pathology itself. Rather,
mice treated with phenylbutyrate displayed elevated H4
acetylation accompanied by an increased production of
proteins implicated in synaptic function
[29] . Therefore,
the use of HDACis has emerged as a promising new strat-
egy for the therapeutic intervention of neurodegenerative
disease
[30 –32] .
Which HDAC is responsible for the cognitive en-
hancement observed following HDACi treatment? To ad-
dress this question, we used a combination of pharmaco-
logical and mouse genetic approaches that allowed us to
identify HDAC2 as a specific negative regulator of synap-
tic plasticity and memory formation
[4] . Chromatin im-
munoprecipitation experiments revealed that HDAC2
associates with the promoters, and likely suppresses the
expression, of a number of genes whose products are im-
plicated in synapse formation/remodeling and memory
formation
[4] . These results suggest that HDACis en-
hance cognitive function by inhibiting HDAC2, render-
ing HDAC2 target genes more accessible to transcription-
al activators and coactivators recruited by neuronal ac-
tivity stimulation. In addition, the impaired cognitive
phenotype of HDAC2-overexpressing mice is readily cor-
rected by SAHA; however, the HDAC2 KO mice are com-
pletely refractory to SAHA treatment. Thus, HDAC2 is
likely to be the major target underlying the beneficial ef-
fects of HDACi treatment. The data presented at the 2011
Barcelona ADPD Conference delineate a novel and im-
portant role for HDAC2 activity in the cognitive impair-
ments associated with neurodegenerative disease.
Conclusion
Our findings delineate an epigenetic mechanism that
underlies memory impairment in neurodegeneration.
These discoveries provide hope for the beneficial effects
of HDAC inhibition. In our model ( fig.1 ), HDAC2 occu-
pancy on the promoters of genes implicated in learning
and memory inhibits the expression of these genes and
The Role of Epigenetic Mechanisms Neurodegenerative Dis
3
leads to cognitive impairment. The recovery of histone
acetylation, via HDAC inhibition, restores histone acety-
lation and the expression of learning and memory and
synaptic plasticity-related genes. The restoration of this
gene expression leads to a reversal of memory deficits.
These data are exciting because they provide hope for
the treatment of neurodegenerative diseases, such as AD,
even when the disease is relatively advanced. Recent clin-
ical trials of pharmacological agents targeting the amy-
loid precursor protein processing pathway in AD have
not provided positive outcomes
[33–37] . One reason giv-
en for this has been that the patients chosen for the trial
were too advanced in their disease, and that these thera-
pies would be most beneficial in people prior to the onset
of AD. However, an estimated 35 million people world-
wide are currently living with dementia, of which AD is
the most common cause
[38 4 0] . Should we focus our ef-
forts entirely on preventative or pre-disease stage thera-
peutics and leave these people to their fate? We believe
that a focus upon treatments aimed at ameliorating the
cognitive symptoms and memory loss of dementia, even
when neuronal loss has already occurred, is equally im-
portant in improving the treatment options for people
with neurodegenerative disease. In this realm, therapeu-
tics targeting chromatin remodeling enzymes such as the
HDACs hold great promise.
HDACis facilitate neuronal
activity-induced gene
expression
Activity-regulated genes: BDNF, c-Fos,
others
Synaptic remodeling/formation: SVP,
Nrxns, SHANKs, others
Synaptic plasticity: NR2A/2B,
GluR1/2, CREB, CBP, CaMKII, others
HDACi treatment
X
Color versi on available online
Fig. 1. HDAC2 regulates learning and
memory by suppressing neuronal gene ex-
pression. HDAC2 is present in transcrip-
tional repressor complexes containing
CoREST and is present on the promoters
of activity-regulated genes. HDAC2 ap-
pe ars to be t he mai n tar get of HDAC is t hat
facilitate memory formation by de-re-
pressing HDAC2 target genes. HDACi
treatment can lead to increased histone
acetylation locally, resulting in relaxed
chromatin structure and increased acces-
sibility for transcr iption activation factors,
such as CREB/CBP complexes. The tran-
scriptional activation of CREB/CBP may
further facilitate the expression of neuro-
nal genes functioning in synaptic plastici-
ty, learning and memory, in part through
its associated histone acetyltransferase ac-
tivity. Adapted from Guan et al. [4].
References 1 Kazantsev AG, Thompson LM: Therapeutic
application of histone deacetylase inhibitors
for central nervous system disorders. Nat
Rev Drug Discov 2008;
7: 854–868.
2 Levenson JM, O’Riordan KJ, Brown KD, et
al: Regulation of histone acetylation during
memory formation in the hippocampus. J
Biol Chem 2004;
279: 40545–40559.
3 Pittenger C, Kandel ER: In search of general
mechanisms for long-lasting plast icity: aply-
sia and the h ippocampus. Phi los Trans R So c
B Biol Sci 2003;
358: 757–763.
4 Guan JS, Haggarty SJ, Giacometti E, et al:
HDAC2 negatively regu lates memory form a-
tion and synaptic plasticity. Nature 2009;
459: 55–60.
5 Brow nell JE, Allis CD: Spec ial HATs for spe-
cial occasions: linking histone acetylation
to chromatin assembly and gene activation.
Curr Opin Genet Dev 1996;
6: 176–18 4.
6 Bird A: Perceptions of epigenetics. Nature
2007;
447: 396–398.
Mungenast /Tsai
Neurodegenerative Dis
4
7 Berger SL , Kouzarides T, Shiek hattar R, et a l:
An operational definition of epigenetics.
Genes Dev 2009;
23: 781–783.
8 de Ru ijter AJ, van Gennip AH, Caron HN, et
al: Histone deacetylases (HDACs): charac-
terization of the cla ssical HDAC family. Bio-
chem J 2003;
370: 737–749.
9 Peleg S, Sananbenesi F, Zovoilis A, et al:
Altered histone acetylation is associated
with age-dependent memory impairment
in mice. Science 2010;
328: 753–756.
10 Salisbur y CM, Cravatt BF: Activ ity-based
probes for proteomic prof iling of histone
deacetylase complexes. Proc Natl Acad Sci
USA 2007;
104: 1171–1176 .
11 Baumann K: Abnormal Alzheimer-like
phosphoryl ation of tau-protein by cycli n-de-
pendent kinases cd k2 and cdk5. FEBS Lett
1993;
336: 417–424.
12 Paudel HK, Lew J, Ali Z, et al: Bra in proline-
directed protein kinase phosphorylates tau
on sites that are abnormally phosphorylated
in tau ass ociated with A lzheimer’s paired he-
lical f ilaments. J Biol Chem 1993;
268: 23512–
23518.
13 Aug ustinack JC , Sanders JL, Tsai L , et al: Co-
localization and fluorescence resonance en-
ergy transfer between cd k5 and AT8 sug-
gests a close association in pre-neurofibril-
lary tangles and neurofibrillary tangles. J
Neuropathol Exp Neurol 2002;
61: 557–564.
14 Pei JJ, Grundke-Iqba l I, Iqbal K, et al: Ac-
cumulation of cyclin-dependent kinase 5
(cdk5) in neurons with early stages of Alz-
heimer’s disease neurofibrillary degenera-
tion. Brain Res 1998;
797: 267–277.
15 Tsai LH, Delalle I, Cav iness VS, et al: p35 is a
neural-specif ic regulatory subunit of cyclin-
dependent kinase 5. Nature 1994;
371: 419–
423.
16 Ko J, Humbert S, Bronson RT, et al: p35 and
p39 are essent ial for cyclin- dependent kina se
5 func tion during neuro development. J Neu-
rosci 2001;
21: 6758–6771.
17 Patrick GN, Zukerberg L, Nikolic M, et al:
Conversion of p35 to p25 deregu lates Cdk5
activity and promotes neurodegeneration.
Nature 1999;
402: 615–622.
18 Tseng HC, Zhou Y, Shen Y, et al: A survey of
Cdk5 activator p35 and p25 levels in Alz-
heimer’s disease brains. FEBS Lett 2002;
523:
58–62.
19 Mungenast AE , Tsai LH: Addressing t he
complex etiolog y of Alzheimer’s disease: the
role of p25/Cd k5. Futu re Neurol 2011;
6: 481
496.
20 Lee MS, Kwon YT, Li M, et al: Neurotoxicit y
induces cleavage of p35 to p25 by calpain.
Nature 2000;
405: 360–364.
21 Nguyen MD, Lariviere RC, Julien JP: Dereg-
ulation of Cdk5 in a mouse model of ALS:
toxicity alleviated by perikaryal neurofila-
ment inclusions. Neuron 2001;
30: 135–147.
22 Smit h PD, Crocker SJ, Jac kson-Lewis V, et al:
Cyclin-dependent kinase 5 is a mediator of
dopaminergic neuron loss in a mouse model
of Parkinsons disease. Proc Natl Acad Sci
USA 2003;
100: 13650–13655.
2 3 P aol et ti P, V il a I, Ri fe M , et al : Do pam in erg ic
and glutamatergic signaling crosstalk in
Huntington’s disease neurodegeneration:
the role of p25/cyclin-dependent kinase 5. J
Neurosci 2008;
28: 1009 0–10101.
24 Sen A, Thom M, Mar tinian L, et al: Deregu-
lation of cdk5 in hippocampal sclerosis. J
Neuropathol Exp Neurol 2006;
65: 55–66.
25 Cru z JC, Tseng HC, Goldma n JA, et al: Aber-
rant Cdk5 activation by p25 triggers patho-
logical events leading to neurodegeneration
and neurofibrillary tangles. Neuron 2003;
40: 471–483.
26 Fischer A , Sananbenesi F, Pang PT, et al: O p-
posing roles of transient and prolonged ex-
pression of p25 in synaptic plasticity and
hippocampus-dependent memory. Neuron
2005;
48: 825–838.
27 Fischer A, Sananbenesi F, Wang X, et al: Re-
cove ry of le arni ng and m emory is asso ciated
with chromatin remodelling. Nature 2007;
447: 178–182 .
28 Kilgore M, Miller CA, Fass DM, et al: Inhib-
itors of class 1 histone deacetylases reverse
contextu al memory defic its in a mouse mod-
el of Alzheimer’s diseas e. Neuropsychophar-
macology 2009;
35: 870–880.
29 Ricobaraza A, Cuadrado-Tejedor M, Perez-
Mediavilla A, et al: Phenylbutyrate amelio-
rates cognitive def icit and reduces tau pa-
thology in an Alzheimer’s disease mouse
model. Neurops ychopharmacology 20 09;
34:
1721–1732.
3 0 C hua ng D M, L eng Y, Ma ri nov a Z, et a l: M ul-
tiple roles of HDAC inhibition in neuro-
degenerative conditions. Trends Neurosci
2009;
32: 591–601.
31 Mai A, Rotili D, Valente S, et al: Histone
deacetylase inhibitors a nd neurodegenera-
tive disorders: holding the promise. Curr
Pharm Des 2009;
15: 3940 –3957.
32 Selvi BR, Cassel JC, Kundu TK, et al: Tuning
acetylation levels w ith HAT activators: ther-
apeutic strategy in neurodegenerative dis-
eases. Bio chim Biophys Acta 2010;
1799: 840–
853.
33 Gauthier S, Aisen PS, Ferris SH, et al: Effect
of tramiprosate in patients with mild-to-
moderate Alzheimer’s disease: exploratory
analyses of the MRI sub-group of the Al-
phase study. J Nutr Health Aging 2009;
13:
550–557.
34 Saumier D, Duong A, Haine D, et al: Do-
main-specific cognitive effects of tramipro-
sate in patients with mild to moderate Alz-
heimer’s disease: ADAS-cog subscale results
from the A lphase study. J Nutr Healt h Aging
2009;
13: 808–812.
35 Holmes C, Boche D, Wilkinson D, et al:
Long-term effects of Abeta42 immunisation
in Alzheimer’s disease: follow-up of a ran-
domised, placebo-controlled phase I trial.
Lancet 2008;
372: 216–223.
36 Patton RL, Kalback WM, Esh CL, et al: A m-
yloid-beta peptide remnants in AN-1792-
immunized Alzheimer’s disease patients: a
biochemica l analysis. Am J Pathol 2006;
169:
1048–1063.
37 Tala n J: High-dose bapineu zumab puts some
AD patients at risk for vasogenic cerebral
edema: those taking largest dose removed
from trials. Neurol Today 2009;
9: 4.
38 Alzheimer’s Disease International. World
Alzheimer report 2010: the global economic
impact of dementia. London, Alzheimer’s
Disease International: The Internationa l
Federation of Alzheimer’s Disease and Re-
lated Disorders Societies, 2010.
39 Alzheimer’s Disease International. World
Alzheimer report 2009: the global economic
impact of dementia. London, Alzheimer’s
Disease International: The Internationa l
Federation of Alzheimer’s Disease and Re-
lated Disorders Societies, 2009.
40 Hardy J: A hu ndred years of Alz heimer’s dis-
ease research. Neuron 2006;
52: 3–13.
© Free Author
Copy – for per-
sonal use only
ANY DISTRIBUTION OF THIS
ARTICLE WITHOUT WRITTEN
CONSENT FROM S. KARGER
AG, BASEL IS A VIOLATION
OF THE COPYRIGHT.
Written permission to distrib-
ute the PDF will be granted
against payment of a per-
mission fee, which is based
on the number of accesses
required. Please contact
permission@karger.ch
© Free Author Copy – for per sonal use only
ANY DISTRIBUTION OF THIS ARTICLE WITHOUT WRITTEN CONSENT FROM S. KARGER AG, BASEL IS A VIOLATION OF THE COPYRIGHT.
Written permission to distribute the PDF will be granted against payment of a per mission fee, which is based on the number of accesses required. Please contact permission@karger.ch
... It was shown that HDACs are involved in the regulation of synaptic plasticity and long term memory formation (Alarson et al., 2004;Korzus et al., 2004). As was discovered in the past few years, different HDACs are selectively involved in the formation of different types of conditioned and unconditioned reflexes (Guan et al., 2009;Bahari Javan et al., 2012;Mungenast and Tsai, 2012;Sando et al., 2012). ...
... Furthermore, their activation is regulated through dif ferent intracellular regulatory systems, which must be activated synchronously (Alberini, 2009;Grinkevich, 2012a). Vol. 4 No. 6 2014 With regard to the long term memory, the recent studies have shown that different HDACs are selec tively involved in the formation of different types of conditioned and unconditioned reflexes (Guan et al., 2009;Mungenast and Tsai, 2012;Bahari Javan et al., 2012). This specificity can be explained by both the differences in their corepressor complexes and differ ent expression profiles of HDACs in functionally dif ferent neurons (Takase et al., 2013). ...
Article
Full-text available
The major tasks of neurobiology include the understanding of mechanisms governing long-term memory formation and search for means to improve memory. Animals with dysfunction of the serotonergic system are a convenient model for investigation of memory processes. The ablation of serotonergic neurons by the neurotoxin 5,7-DOT leads to the inability of Helix snails to form an aversive food avoidance reflex. Previously we have found that epigenetic processes, such as histone methylation and acetylation, are involved in the formation of food aversion, and that disturbance of these processes leads to the inability to form long-term memory. The goal of this study was to investigate the possibility to reverse long-term memory in DOT-treated animals through the induction of acetylation processes. We found that treatment with histone deacetylase inhibitors NaB and Trichostatin A significantly increased the ability of DOT-treated animals to form the food aversion reflex. The results point to an important role of serotonin in the induction of the epigenetic processes mediating the formation of this type of long-term memory and to significantly improve its parameters by induction of acetylation processes. Our Neurodegeneration model, based on ablation of serotonergic neurons, can be useful in studies of the epigenetic mechanisms underlying long-term memory destruction and screening of compounds crucial for memory formation.
... In early injury, as the brain attempts to adapt to the trauma, a broad genomic, molecular and morphologic reorganization in neuronal and glial activity relationships occurs in response to the injury. Some of these changes result in permanent inhibition of neuronal activity in both affected and intact parts of the brain [30,31]. Therefore, the early administration of progesterone in TBI may cause the recovery of the patients. ...
Article
Full-text available
Background and objective: The benefits of progesterone have been demonstrated in the animal models of traumatic brain injury (TBI). However, the results of clinical studies are conflicting. Considering the heterogenic nature of TBI, the effect of progesterone in patients with diffuse axonal injury (DAI) was investigated in a clinical trial. Methods: In this study, 48 patients with DAI and Glasgow Coma Scale of 3–12, admitted within 4 hours after injury, were randomly assigned to the progesterone or control group. The dose of progesterone administration was 1 mg kg–1 per 12 hours for 5 days. The effect of progesterone was investigated using extended-Glasgow Outcome Scale (GOS-E), functional independence measure (FIM) scores and also mortality within the follow-up period. Results: The progesterone group exhibited higher GOS-E and FIM scores in comparison to the control group at 6 months post-injury (p < 0.01 and p < 0.05, respectively). Mortality was also found in the control group (p < 0.05). The adverse events attributed to the progesterone administration were not found throughout the study. Conclusions: Findings of this study suggest that progesterone may be neuroprotective in patients with DAI. However, large clinical trials are needed to assess progesterone as a promising drug in DAI.
... Guan et al. then demonstrated that overexpression of HDAC2 led to decreased dendritic spine density [5]. Because histone acetylation status is altered during memory formation, HDACIs have recently gained attention in the field of neurodegenerative diseases [6]. Transcriptionally active chromatin structure has been shown to be crucial for functional learning and memory in mouse neurons, and the activity of chromatin structure depends on epigenetic mechanisms such as histone acetylation [7]. ...
Article
Background: The accumulation of amyloid-β (Aβ) leads to the loss of dendritic spines and synapses, which is hypothesized to cause cognitive impairments in Alzheimer's disease (AD) patients. In our previous study, we demonstrated that a novel mercaptoacetamide-based class II histone deacetylase inhibitor (HDACI), known as W2, decreased Aβ levels and improved learning and memory in mice. However, the underlying mechanism of this effect is unknown. Objective: Because dendritic spine formation is associated with cognitive performance, here we investigated whether HDACI W2 regulates dendritic spine density and its molecular mechanism of action. Methods: To examine the effect of HDACI W2 on dendritic spine density, we conducted morphological analysis of dendritic spines using GFP transfection and Golgi staining. In addition, to determine the molecular mechanism of W2 effects on spines, we measured the levels of mRNAs and proteins involved in the Ras signaling pathway using quantitative real-time PCR, immunocytochemistry, and western analysis. Results: We found that HDACI W2 altered dendritic spine density and morphology in vitro and in vivo. Additionally, W2 increased the mRNA or protein levels of Ras GRF1 and phospho-ERK. Moreover, knockdown of RasGRF1 and inhibition of ERK activity prevented the W2-mediated spinogenesis in primary hippocampal neurons. Conclusion: Our Class II-selective HDACI W2 promotes the formation and growth of dendritic spines in a RasGRF1 and ERK dependent manner in primary hippocampal neurons.
... As such, stress-induced behaviors are not reflective of human MDD per se, but represent, at best, aspects associated with the human disorder. Although Cdk5 expression has been suggested to mediate stress responses 14 and contribute to epigenetic programing 26 , it has not been linked genetically to MDD or other mental illnesses, likely as a result of its essential role in many neuronal and developmental functions. Genome-wide association studies (GWAS) of MDD are in early phases and do not appear to have yet positively identified or confirmed risk factors for depression. ...
Article
The cAMP and cAMP-dependent protein kinase A (PKA) signaling cascade is a ubiquitous pathway acting downstream of multiple neuromodulators. We found that the phosphorylation of phosphodiesterase-4 (PDE4) by cyclin-dependent protein kinase 5 (Cdk5) facilitated cAMP degradation and homeostasis of cAMP/PKA signaling. In mice, loss of Cdk5 throughout the forebrain elevated cAMP levels and increased PKA activity in striatal neurons, and altered behavioral responses to acute or chronic stressors. Ventral striatum- or D1 dopamine receptor-specific conditional knockout of Cdk5, or ventral striatum infusion of a small interfering peptide that selectively targeted the regulation of PDE4 by Cdk5, produced analogous effects on stress-induced behavioral responses. Together, our results demonstrate that altering cAMP signaling in medium spiny neurons of the ventral striatum can effectively modulate stress-induced behavioral states. We propose that targeting the Cdk5 regulation of PDE4 could be a new therapeutic approach for clinical conditions associated with stress, such as depression.
... Areas both proximal and distal to the locus of the injury are deafferented, and latent pathways are unmasked which can activate or inhibit neuronal areas, leading to potentially maladaptive structural and functional changes that would not occur in the intact brain. Some of these changes result in permanent inhibition of neuronal activity in both the affected and intact parts of the brain (diaschisis) (Nishibe et al., 2010;Dancause and Nudo, 2011;Mungenast and Tsai, 2012;see Stein, 2012, for discussion). These very diffuse changes in brain structure can produce behavioral deficits and impairments that can eventually result in permanent dysfunction due to hypo-as well as hyperactivity induced by neurotransmitter-modifying drugs. ...
Chapter
The clinical understanding of traumatic brain injury (TBI) and its manifestations is beginning to change. Both clinicians and research scientists are recognizing that TBI and related disorders such as stroke are complex, systemic inflammatory and degenerative diseases that require an approach to treatment more sophisticated than targeting a single gene, receptor, or signaling pathway. It is becoming increasingly clear that TBI is a form of degenerative disorder affecting the brain and other organs, and that its manifestations can unfold days, weeks, and years after the initial damage. Until recently, and despite numerous industry- and government-sponsored clinical trials, attempts to find a safe and effective neuroprotective agent have all failed - probably because the research and development strategies have been based on an outdated early 20th century paradigm seeking a magic bullet that will affect a narrowly circumscribed target. We propose that more attention be given to the development of drugs, given alone or in combination, that are pleiotropic in their actions and that have systemic as well as central nervous system effects. We review current Phase II and Phase III trials for acute pharmacologic treatments for TBI and report on their aims, methods, status, and important associated research issues. © 2015 Elsevier B.V. All rights reserved.
Article
Neuroinflammation in the hippocampus plays essential roles in postoperative cognitive dysfunction (POCD). Histone deacetylases (HDACs) have recently been identified as key regulators of neuroinflammation. MS-275, an inhibitor of HDAC, has been reported to have neuroprotective effects. Therefore, the present study aimed to test the hypothesis that pretreatment with MS-275 prevents POCD by inhibiting neuroinflammation in rats. In this study, anesthesia/surgery impaired cognition, demonstrated by an increase escape latency and reduction in the number of platform crossings in Morris water maze (MWM) trials, through activating microglia neuroinflammation and decreasing PSD-95 expression. However, pretreatment with MS-275 attenuated postoperative cognitive impairment severity. Furthermore, pretreatment with MS-275 decreased activated microglia levels and increased PSD95 protein expression in the hippocampus. Pretreatment with MS-275 reduced NF-κB-p65 protein expression and nuclear accumulation as well as the neuroinflammatory response (production of proinflammatory cytokines including TNF-α and IL-1β) in the hippocampus. Additionally, MS-275 reduced HDAC2 expression and HDAC activity in the hippocampus, which were enhanced in vehicle-treated rats. These results suggest that MS-275 alleviates postoperative cognitive dysfunction by reducing neuroinflammation in the hippocampus of rats via HDAC inhibition.
Article
Aberrant changes in gene function are believed to be involved in a wide spectrum of human disease including behavioral, cognitive and neurodegenerative pathologies. Most of the attention in the last few decades has focused on changes in gene sequence as a cause of gene dysfunction leading to disease and mental health disorders. Germ line mutations or other alterations in the sequence of DNA that associate with different behavioral and neurological pathologies have been identified. However, sequence alterations explain only a small faction of the cases. In addition there is evidence for “gene- environment” interactions in the brain suggesting mechanisms that alter gene function and the phenotype through environmental exposure. Genes are programmed by “epigenetic” mechanisms such as chromatin structure, chromatin modification and DNA methylation. These mechanisms confer on similar sequences different identities during cellular differentiation. Epigenetic differences are proposed to be involved in differentiating gene function in response to different environmental contexts and could result in alterations in functional gene networks that lead to brain disease. Epigenetic markers could serve important biomarkers in brain and behavioral diseases. Moreover, epigenetic processes are potentially reversible pointing to epigenetic therapeutics in psychotherapy.
Article
Full-text available
Chromatin modifications, especially histone-tail acetylation, have been implicated in memory formation. Increased histone-tail acetylation induced by inhibitors of histone deacetylases (HDACis) facilitates learning and memory in wild-type mice as well as in mouse models of neurodegeneration. Harnessing the therapeutic potential of HDACis requires knowledge of the specific HDAC family member(s) linked to cognitive enhancement. Here we show that neuron-specific overexpression of HDAC2, but not that of HDAC1, decreased dendritic spine density, synapse number, synaptic plasticity and memory formation. Conversely, Hdac2 deficiency resulted in increased synapse number and memory facilitation, similar to chronic treatment with HDACis in mice. Notably, reduced synapse number and learning impairment of HDAC2-overexpressing mice were ameliorated by chronic treatment with HDACis. Correspondingly, treatment with HDACis failed to further facilitate memory formation in Hdac2-deficient mice. Furthermore, analysis of promoter occupancy revealed an association of HDAC2 with the promoters of genes implicated in synaptic plasticity and memory formation. Taken together, our results suggest that HDAC2 functions in modulating synaptic plasticity and long-lasting changes of neural circuits, which in turn negatively regulates learning and memory. These observations encourage the development and testing of HDAC2-selective inhibitors for human diseases associated with memory impairment.
Article
Full-text available
Alzheimers disease (AD) is an age-related neurodegenerative disorder characterized by the progressive loss of forebrain neurons and the deterioration of learning and memory. Therapies for AD have primarily focused upon either the inhibition of amyloid synthesis or its deposition in the brain, but clinical testing to date has not yet found an effective amelioration of cognitive symptoms. Synaptic loss closely correlates with the degree of dementia in AD patients. However, mouse AD models that target the amyloid-β pathway generally do not exhibit a profound loss of synapses, despite extensive synaptic dysfunction. The increased generation of p25, an activator of the cyclin-dependent kinase 5 (Cdk5) has been found in both human patients and mouse models of neurodegeneration. The current work reviews our knowledge, to date, on the role of p25/Cdk5 in Alzheimers disease, with a focus upon the interaction of amyloid-β and p25/Cdk5 in synaptic dysfunction and neuronal loss.
Article
Full-text available
Immunisation of patients with Alzheimer's disease with full-length amyloid-beta peptide (Abeta(42)) can clear amyloid plaques from the brain. Our aim was to assess the relation between Abeta(42) immune response, degree of plaque removal, and long-term clinical outcomes. In June, 2003, consent for long-term clinical follow-up, post-mortem neuropathological examination, or both, was sought from 80 patients (or their carers) who had entered a phase I randomised, placebo-controlled trial of immunisation with Abeta(42) (AN1792, Elan Pharmaceuticals) in September, 2000. The follow-up study was completed in September, 2006. Plaques were assessed in terms of the percentage area of the cortex with Abeta immunostaining (Abeta load) and in terms of characteristic histological features reflecting plaque removal. Survival of all 80 individuals until severe dementia or death was assessed with a Cox proportional hazard model. 20 participants--15 in the AN1792 group, five in the placebo group--died before follow-up started. A further 22 patients--19 in the AN1792 group, three in the placebo group--died during follow-up. Nine of the deceased patients, all in the AN1792 group, had given consent for post-mortem analysis; one of these who did not die with Alzheimer's disease was excluded. In the remaining eight participants who received immunisation and who were examined neuropathologically, mean Abeta load was lower than in an unimmunised control group that was matched for age at death (2.1% [SE 0.7] in treated participants vs 5.1% [0.9] in controls; mean difference 3.0%, 95% CI 0.6-5.4; p=0.02). Although there was considerable variation in Abeta load and degree of plaque removal among immunised participants, the degree of plaque removal varied significantly with mean antibody response attained during the treatment study period (Kruskal-Wallis p=0.02). Seven of the eight immunised patients who underwent post-mortem assessment, including those with virtually complete plaque removal, had severe end stage dementia before death. In the whole cohort, there was no evidence of improved survival (hazard ratio 0.93, 95% CI 0.43-3.11; p=0.86) or of an improvement in the time to severe dementia (1.18, 0.45-3.11; p=0.73) in the AN1792 group versus the placebo group. Although immunisation with Abeta(42) resulted in clearance of amyloid plaques in patients with Alzheimer's disease, this clearance did not prevent progressive neurodegeneration.
Article
Full-text available
As the human life span increases, the number of people suffering from cognitive decline is rising dramatically. The mechanisms underlying age-associated memory impairment are, however, not understood. Here we show that memory disturbances in the aging brain of the mouse are associated with altered hippocampal chromatin plasticity. During learning, aged mice display a specific deregulation of histone H4 lysine 12 (H4K12) acetylation and fail to initiate a hippocampal gene expression program associated with memory consolidation. Restoration of physiological H4K12 acetylation reinstates the expression of learning-induced genes and leads to the recovery of cognitive abilities. Our data suggest that deregulated H4K12 acetylation may represent an early biomarker of an impaired genome-environment interaction in the aging mouse brain.
Article
Full-text available
Alzheimer's disease (AD) is a neurodegenerative disorder characterized clinically by cognitive impairments that progress to dementia and death. The earliest symptoms of AD present as a relatively pure deficit in memory retrieval. Therefore, drug treatments that intervene in the early stages of AD by rescuing memory deficits could be promising therapies to slow, or even reverse progression of the disease. In this study, we tested the potential of systemic histone deacetylase inhibitor (HDACi) treatment to rescue cognitive deficits in a mouse model of AD. APPswe/PS1dE9 mice showed pronounced contextual memory impairments beginning at 6 months of age. Chronic HDACi injections (2-3 weeks) did not alter contextual memory formation in normal mice, but had profound effects in transgenic animals. Injections of sodium valproate, sodium butyrate, or vorinostat (suberoylanilide hydroxamic acid; Zolinza) completely restored contextual memory in these mutant mice. Further behavioral testing of the HDACi-treated transgenic mice showed that the newly consolidated memories were stably maintained over a 2-week period. Measurement of the HDAC isoform selectivity profile of sodium valproate, sodium butyrate, and vorinostat revealed the common inhibition of class I HDACs (HDAC1, 2, 3, 8) with little effect on the class IIa HDAC family members (HDAC4, 5, 7, 9) and inhibition of HDAC6 only by vorinostat. These preclinical results indicate that targeted inhibition of class I HDAC isoforms is a promising avenue for treating the cognitive deficits associated with early stage AD.
Article
Cyclin-dependent kinase 5 (Cdk5) is required for proper development of the mammalian central nervous system. To be activated, Cdk5 has to associate with its regulatory subunit, p35. We have found that p25, a truncated form of p35, accumulates in neurons in the brains of patients with Alzheimer's disease. This accumulation correlates with an increase in Cdk5 kinase activity. Unlike p35, p25 is not readily degraded, and binding of p25 to Cdk5 constitutively activates Cdk5, changes its cellular location and alters its substrate specificity. In vivo the p25/Cdk5 complex hyperphosphorylates tau, which reduces tau's ability to associate with microtubules. Moreover, expression of the p25/Cdk5 complex in cultured primary neurons induces cytoskeletal disruption, morphological degeneration and apoptosis. These findings indicate that cleavage of p35, followed by accumulation of p25, may be involved in the pathogenesis of cytoskeletal abnormalities and neuronal death in neurodegenerative diseases.
Article
An abstract is unavailable. This article is available as HTML full text and PDF.
Article
The efficacy, safety and disease-modification of tramiprosate (homotaurine)were investigated in a recently completed large-scale Phase III clinical study in patients with mild to moderate Alzheimer's disease (AD), the Alphase study. Disease-modification was assessed using longitudinal volumetric MRI (vMRI) measurements of the hippocampus in a subgroup of patients. The present study describes the vMRI, cognitive and clinical results obtained in this subgroup. Multi-center, double-blind, randomized, placebo-controlled study in a subset of the 1052 patients of the Alphase study. Setting: 51 vMRI investigative sites in the United States and Canada. A total of 508 patients underwent vMRI scanning. Of these, 312 provided scan pairs for assessing hippocampus volume changes and were included in the analyses. Patients were randomized to receive Placebo BID (n = 109), tramiprosate 100 mg BID (n = 103), or tramiprosate 150 mg BID (n = 100) for 78 weeks. Alzheimer's Disease Assessment Scale-cognitive subscale (ADAS-cog) and Clinical Dementia Rating-Sum-of-boxes CDR-SB assessments were conducted at Baseline and at Weeks 13, 26, 39, 52, 65 and 78. Exploratory analyses were performed using similar First and Final mixed-effects repeated-measures models that were used for the analysis of the entire patient dataset. Psychometric score results showed numerical trends in favour of tramiprosate that did not reach statistical significance. While there were no statistically significant group differences in hippocampus volume using the First modeling approach, a significant dose-response reduction in hippocampus volume change was found in the Final models. Moreover, there was a marginally significant overall treatment main effect and a significant slope difference in favour of tramiprosate according to the Final model analysis of the ADAS-cog scores. ADAS-cog scores analyzed according to this model also revealed differences in favor of the tramiprosate 150 mg group at weeks 26 and 52, with marginally significant differences at Weeks 13 and 39. Slope analyses of ADAS-cog score changes showed significant differences in favor of the 150 mg BID group, and when both active groups were combined, in comparison to the placebo group. No between-group differences with respect to changes to each visit in the CDR-SB were observed with either modeling approach. Although there was a similar dose-response relationship observed in the hippocampus volume and ADAS-cog Final model analyses, the overall changes in psychometric scores and hippocampus volume were not significantly correlated. Exploratory analysis of the vMRI subgroup suggests that tramiprosate slows hippocampal atrophy, and reveals some evidence of a beneficial effect on cognition. The clinical validity of the vMRI biomarker is discussed.
Article
Neurodegenerative diseases, such as polyglutamine-related diseases, amyotrophic lateral sclerosis, and Alzheimer's disease are accompanied by transcriptional dysfunctions, leading to neuronal death. It is becoming more evident that the chromatin acetylation status is impaired during the lifetime of neurons, by a common mechanism related to the loss of function of histone acetyltransferase (HAT) activity. Notably, the HAT termed cAMP response element binding protein (CREB)-binding protein (CBP) was shown to display neuroprotective functions. Several other HATs have now been shown to participate in basic but vital neuronal functions. In addition, there is increasing evidence of several HATs (including CBP), as essential regulators of neuronal plasticity and memory formation processes. In order to counteract neuronal loss and/or memory deficits in neurodegenerative diseases, the current therapeutic strategies involve the use of small molecules antagonizing histone deacetylase (HDAC) activity (i.e. HDAC inhibitors). Although this strategy lacks specificity, some of these molecules display promising therapeutic properties. With the rapidly evolving literature on HATs and their respective functions in neuronal survival and memory formation, it seems essential to envisage direct stimulation of the acetyltransferase function as a new therapeutic tool in neurodegenerative diseases. In this review, we will highlight the present understanding and the future prospects of such therapeutic approach.