Abstract
One of the main focuses in Huntington's disease (HD) research, as well as in most neurodegenerative diseases, is the development of new therapeutic strategies, as currently there is no treatment to delay or prevent the progression of the disease. Neuronal dysfunction and neuronal death in HD are caused by a combination of interrelated pathogenic processes that lead to motor, cognitive and psychiatric symptoms. Understanding how mutant huntingtin impacts on a plethora of cellular functions could help to identify new molecular targets. Although HD has been classically classified as a neurodegenerative disease affecting voluntary movement, lately cognitive dysfunction is receiving increased attention as it is very invalidating for patients. Thus, an ambitious goal in HD research is to find altered molecular mechanisms that contribute to cognitive decline. In this review, we have focused on those findings related to corticostriatal and hippocampal cognitive dysfunction in HD, as well as on the underlying molecular mechanisms, which constitute potential therapeutic targets. These include alterations in synaptic plasticity, transcriptional machinery and neurotrophic and neurotransmitter signaling.
Keywords: A2AR, cGMP, GluN3A, Kalirin‐7, nSOC, PDEs, PKA, p75NTR
INTRODUCTION
In Huntington's disease (HD), the trinucleotide (CAG) repeat expansion in the huntingtin gene results in an extended polyglutamine (polyQ) tract in the huntingtin protein, which induces a cascade of pathological changes leading to neuronal dysfunction and neurodegeneration. HD is characterized by degeneration of the striatum and cortical atrophy 246, 321, but other brain areas such as the hippocampus, thalamus, globus pallidus and substantia nigra are also affected 117, 124, 134, 199, 229, 249, 285, 321. Importantly, evidence from both patients 40 and mouse models 158, 305 indicates that cell death does not occur until late stages of the disease, indicating that neuronal dysfunction, including abnormal synaptic plasticity, is an early pathogenic event that precedes neuronal death and leads to HD symptoms. Having this in mind, research is focused on finding common molecular targets to prevent cognitive impairment by preserving/restoring neuronal function at early disease stages, which hopefully will help to avoid subsequent neuronal death. Thus, the identification of these early cellular and molecular pathogenic events in HD represents an important milestone in the design of new therapeutic approaches to cure or delay disease progression. To achieve this goal it is mandatory to build from data obtained by analyzing HD mouse models, as new therapeutic strategies can be developed based on those altered molecular mechanisms found to contribute to cognitive impairment in HD.
COGNITIVE DYSFUNCTION IN HD PATIENTS
The first paper on cognitive function in HD was published in 1974 23, and relevant publications on this issue have emerged at an accelerating rate since then [reviewed by Stout et al. 290], so that the nature and variety of cognitive decline in patients with HD are well documented and have been the focus of recent reviews 72, 113, 217, 291, 317.
Corticostriatal dysfunction
Although HD is characterized by a progressive degeneration of medium‐sized striatal spiny neurons (MSNs) 81, 322, functional and morphological changes in the neocortex have been proposed as the initial triggers of striatal pathology, and it has been put forward that cortical changes are fundamental for the onset and progression of the HD phenotype, in both humans and animal models [reviewed by Estrada‐Sanchez and Rebec 77]. In this context, HD patients at pre‐symptomatic stages exhibit alterations in tasks related to the neocortical function, like those that require strategy shift 119, 151, and also in executive function, in verbal fluency 112, 219, in procedural learning, in planning and in explicit motor learning 112, 149, 252, 270. Moreover, early corticostriatal alterations in HD patients have been further attested by neuroimaging studies showing impaired corticostriatal functional connectivity in pre‐symptomatic HD subjects 234, 235, 307. These corticostriatal alterations correlate with decreased activity in the frontal cortex and putamen 140. In mid‐stage clinically symptomatic HD patients, executive function, verbal fluency, perceptual speed and reasoning are strongly affected 7, 156, while at more advanced disease stages, a sub‐cortical dementia gradually develops, with alterations in several simple and complex cognitive functions involving slow information processing, decreased motivation, depression, apathy and personality changes 218, 346.
Marked nuclear and cytoplasmic accumulation of mutant huntingtin has been reported in cortical pyramidal neurons, which overlaps with cortical dendritic abnormalities in postmortem brain of patients with low‐grade striatal pathology 70, 265. Indeed, abundant mutant huntingtin is evident in numerous cortical dystrophic axons that project to the striatum 264. It has also been reported that stage I and II HD patients exhibit specific thinning of the neocortex 134, 250. Overall, these findings indicate that cortical changes are fundamental to the onset and progression of the HD phenotype, evidencing a central role of the cortex at early stages of HD.
Hippocampal dysfunction
The hippocampus, together with the amygdala and the nucleus accumbens, forms the central axis of the limbic system that plays a key role in the formation of declarative memory, spatial learning and awareness, navigation, object recognition and visual memory, as well as in executive functions 21, 38, 74, 109, 131, 133, 138, 300. Studies in HD patients have mainly focused on the cognitive functions involving the corticostriatal circuitry, whereas those related to hippocampal connectivity are still poorly analyzed. Nevertheless, there is clear evidence of hippocampal morphological alterations in HD patients such as a reduction in hippocampal volume 124, 249, 285 and the presence of mutant huntingtin aggregates 117, 199. Moreover, some cognitive tasks analyzed in human studies such as the evaluation of spatial working memory, spatial recognition memory, object recognition memory, episodic memories and some forms of associative learning, can involve the participation of the hippocampus and temporal lobe structures 38, 54, 75, 109, 138, 196. Thus, although no severe deficits in spatial working memory have been shown in pre‐symptomatic HD patients, their latency in performing these tasks is higher than in control individuals 149, 151, whereas recognition memory has been shown to be affected in pre‐symptomatic HD‐gene carriers 16. A recent study using tests analogous to those employed in HD mouse models describes hippocampal‐dependent impairments in patients in early HD stage, ahead of motor symptoms. Patients are unable to learn the location of the platform in the virtual Morris water maze and performance correlates to the estimated years to disease onset 12. In early‐mild symptomatic HD patients, alterations in associative learning, spatial short‐term memory, spatial working memory and recognition memory have been described 150, 197. Importantly, in middle and more advanced disease stages, a global cognitive decline is observed in patients with HD 7, 156, 218, 346. These alterations involve both corticostriatal and hippocampal dysfunction. However, it seems that declarative memories more related to hippocampal and corticotemporal functions are not as altered as procedural learning, more related to corticostriatal integrity. Actually, in patients with HD, the hippocampus compensates for gradual dysfunction of the caudate nucleus, helping to maintain route recognition close to normal performance 213, 319.
Impaired synaptic plasticity and cognitive dysfunction in HD mouse models
The discovery of the gene mutation responsible for HD occurred in 1993 299, and this allowed for the generation of several HD genetic models. Although the disease has been reproduced in diverse species, mouse models are the most extensively used. The different genetic mouse models differ in their phenotypes as a result of the way mutant huntingtin is inserted into the murine genome: (i) exon‐1 transgenic mice: R6, N171‐82Q and the conditional HD94Q‐tet off mice, and (ii) full‐length transgenic mice: Yeast artificial chromosome (YAC), bacteria artificial chromosome (BAC)HD and knock‐in mice (Table 1). HD mouse models display alterations in synaptic plasticity (Table 2) and replicate the cognitive impairment observed in HD patients (Table 3), thus providing an excellent opportunity to study the underlying molecular mechanisms and to test potential treatments for transfer to patients [for review see Cepeda et al. 43, Chang et al. 49, Menalled and Brunner 187 and Plotkin and Surmeier 228].
Table 1.
Genetically modified HD mouse models.
| Mouse model | Construct | Promoter | CAG repeat size | Lifespan | References | 
|---|---|---|---|---|---|
| R6/1 | Insertion of the exon 1 of human HD gene into mouse genome | Human huntingtin | 116 | 32–40 w | 77, 78, 79, 305 | 
| R6/2 | Insertion of the exon 1 of human HD gene into mouse genome | Human huntingtin | 144 | 13–16 w | 11, 12, 62, 177, 186 | 
| N171‐82Q | Insertion of the first 171 aa from the N‐terminal fragment of the human HD gene into mouse genome | Mouse prion protein | 82 | 16–24 w | 268 | 
| HD94‐ tet off | Insertion of a chimeric mouse/human exon 1 fragment with polyQ expansion into mouse genome | CamKIIa‐tTA | 94 | Normal | 340 | 
| YAC72 | Yeast artificial chromosome expressing full‐length human mutant huntingtin | Human huntingtin | 72 | Normal | 121, 271, 314 | 
| YAC128 | Yeast artificial chromosome expressing full‐length human mutant huntingtin | Human huntingtin | 128 | Normal | 236, 237, 280, 310, 311 | 
| BACHD | Bacterial artificial chromosome expressing full‐length human mutant huntingtin | Human huntingtin | 97 CAG/CAA mixed repeats | Normal | 76, 107, 123, 236 | 
| Hdh92Q | Replacing exon 1 of the mouse huntingtin gene with a mutant human exon 1 | Mouse huntingtin | 92 | Normal | 85, 324, 327 | 
| Hdh111Q | Replacing exon 1 of the mouse huntingtin gene with a mutant human exon 1 | Mouse huntingtin | 111 | Normal | 324, 325, 326, 327 | 
| CAG140 | Inserting CAG repeats into the mouse huntingtin gene | Mouse huntingtin | 140 | Normal | 188 | 
| CAG150 | Inserting CAG repeats into the mouse huntingtin gene | Mouse huntingtin | 150 | Normal | 87, 165, 344 | 
Information about construct insertion, the promoter used to express the mutation, the CAG repeat number and the lifespan. Weeks (w).
Table 2.
Alterations in synaptic plasticity in HD mouse models.
| Mouse model | Cerebral cortex | Striatum | Hippocampus | References | 
|---|---|---|---|---|
| R6/1 | Loss of cortical plasticity (8–10 w) | Decreased spontaneous EPSCs in MSNs (9–13 mo) | Decreased LTP in brain slices (5 w) | 36, 46, 57, 59, 60, 99, 190, 192, 286, 306 | 
| Increased LTD (2 mo) followed by a reduction in LTD expression (5 mo) and loss of LTD (7–9 mo) at perirhinal synapses | Increased spontaneous IPSCs (12–15 mo) | Impaired LTP in vivo (13–14 w) | ||
| Decreased dendritic spine density and spine length (8 mo) | Decreased dendritic spine density and spine length (8 mo) | Increased LTD at CA1 synapses (12 w) | ||
| Decreased dendritic spine density (20 w) | ||||
| R6/2 | Hyperexcitable cortex and greater susceptibility to seizures (3 w) | Progressive decrease in spontaneous EPSCs in MSNs (5–7 w) | Reduced LTP and aberrant LTD at CA1 synapses (5 w) | 4, 45, 47, 57, 58, 115, 122, 141, 148, 166, 200, 201, 254 | 
| Increased spontaneous IPSCs (3–4 w) | No differences in the LF IPSCs | |||
| Decreased spontaneous IPSCs and increased spontaneous EPSCs (13 w) | Increased spontaneous IPSCs (5–7 w and 9–14 w) | |||
| Dendritic spine loss (3–4 w) | Progressive dendritic spine loss (4–10 w) | |||
| Dysfunctional communication between cortex and striatum (7–9 w) | ||||
| YAC72 | Increased NMDAR‐mediated EPSCs in MSNs (21–31 d) after stimulation of cortical afferents in corticostriatal slices | Increased LTP (6 mo) | 121, 161, 162, 194 | |
| Altered early corticostriatal synaptic function (21–30 d), including presynaptic dysfunction and propensity towards synaptic depression | Reduced LTP (10 mo) | |||
| YAC128 | Altered early corticostriatal synaptic function (21–30 d) | Reduced paired‐pulse depression in the DG (3–6 mo) | 57, 58, 90, 129, 179, 331, 334 | |
| Presynaptic dysfunction, propensity towards synaptic depression and altered AMPAR function | Enhanced post‐tetanic and short‐term potentiation after HF stimulation (3–6 mo) | |||
| Biphasic age‐dependent changes in corticostriatal function: | ||||
| Increased synaptic currents and glutamate release (1 mo) | ||||
| Reduced evoked synaptic currents and glutamate release (7–12 mo) | ||||
| Increased spontaneous EPSCs (12 mo) | Reduced spontaneous EPSCs (6 and 12 mo) | |||
| Increase spontaneous IPSCs (6 and 12 mo) | Reduced spontaneous LF IPSCs (6 and 12 mo) | |||
| Increased spontaneous HF IPSCs (1, 2, 12 mo) | ||||
| Progressive dendritic spine density loss (3, 6, 12 mo) | ||||
| BACHD | Progressive reduction of cortical excitation and inhibition of layer 2/3 pyramidal cells (3–6 mo) | Progressive reduction of excitation onto MSNs (3–6 mo) | n.r. | 107, 123, 236, 248, 284 | 
| High excitability of MSNs (18 mo) | ||||
| Reduced dendritic spine density (18 mo) | ||||
| Hdh92Q | n.r. | n.r. | Reduced LTP (4–6 mo) | 172 | 
| Hdh111Q | n.r. | n.r. | Reduced LTP (2 mo) | 172 | 
| Reduced actin polymerization in dendritic spines after TBS‐induced LTP | ||||
| Hdh 7Q/111Q | Impaired induction and maintenance of corticostriatal LTP (2 and 4 mo) | Reduced LTP (6 mo) | 28, 239 | |
| Decreased dendritic spine density and altered distribution with a specific reduction in the proportion of thin spines (8 mo) | ||||
| Decreased dendritic spine density and a shift in their distribution (2 mo) | No alterations in dendritic spine density (2 mo) | |||
| Progressive decrease of glutamatergic excitatory postsynaptic sites (2 and 8 mo) | Decrease of glutamatergic excitatory postsynaptic sites (8 mo) | |||
| CAG140 | Increased spontaneous EPSCs (12 mo) | Reduced spontaneous EPSCs (12 and 18 mo) | n.r. | 57, 58, 66, 157 | 
| Increased spontaneous IPSCs (6 and 12 mo) | Reduced spontaneous LF IPSCs (12 and 18 mo) | |||
| Increased spontaneous HF IPSCs (12 mo) | ||||
| Reduced thalamostriatal axondendritic terminals (1 mo) | ||||
| Loss of corticostriatal terminals (12 mo) | ||||
| Reduced dendritic spines in MSNs (20–26 mo) | ||||
Alterations in synaptic plasticity (electrophysiological properties and dendritic spine density/morphology) of neuronal populations from the cerebral cortex, striatum and hippocampus of HD mouse models. Long‐term depression (LTD); Long‐term potentiation (LTP); Excitatory postsynaptic currents (EPSCs); Inhibitory postsynaptic currents (IPSCs); Cornus Ammonis 1 (CA1); Medium‐sized spiny neurons (MSNs); N‐methyl‐d‐aspartate receptor (NMDAR); α‐amino‐3‐hydroxyl‐5‐methyl‐4‐isoxazole‐propionate receptor (AMPAR); Dentate gyrus (DG); low frequency (LF); high frequency (HF); Theta burst stimulation (TBS); Weeks (w); Months (mo); Not reported (n.r.).
Table 3.
Cognitive, psychiatric and motor deficits in HD mouse models.
| Mouse model | Cognitive and psychiatric deficits | Motor deficits | References | 
|---|---|---|---|
| R6/1 | 8–12 w | 14 w 8 w (AR) | 36, 99, 102, 177, 239 | 
| R6/2 | 4–8 w | 6–8 w | 41, 97, 166, 177, 287 | 
| N171‐82Q | 14 w | 11 w | 184, 243, 268 | 
| HD94‐ tet off | n.r. | 4 w | 69, 340 | 
| YAC72 | n.r. | Hyperactivity (7–9 mo) 16 mo | 121 | 
| YAC128 | 7–8.5 mo | Hyperkinesia (3 mo) | 236, 280, 311, 312, 313 | 
| Hypokinesia (6 mo) | |||
| 2–12 mo (AR) | |||
| BACHD | 2–12 mo | 2–12 mo (AR) | 76, 107, 123, 236 | 
| Hdh92Q | 4 mo | 21 mo | 31, 304 | 
| Hdh111Q | n.r. | 24 mo | 325 | 
| Hdh7Q/111Q | 6 mo | 8 mo | 28, 98, 239, 342, 343 | 
| 2–6 mo (AR) | |||
| CAG140 | 4–6 mo | Hyperkinesia (1–3 mo) | 118, 188 | 
| Hypokinesia (from 3 mo) | |||
| 4–12 mo | |||
| CAG150 | 4 mo | 4–10 mo | 32, 165 | 
Information about the age at which HD models start to exhibit cognitive, psychiatric and motor deficits. Motor deficits include several parameters: 1) learning of new motor skills (evaluated with the accelerating rotarod (AR) task), 2) hypo/hyperactivity, and 3) motor coordination (evaluated using several motor tests). Specific information about each model can be found in the cited references. Weeks (w); Months (mo); Not reported (n.r.).
Electrophysiology is one of the best characterized methods to evaluate synaptic plasticity. Long‐term potentiation (LTP) and long‐term depression (LTD) are commonly used paradigms to assess synaptic properties 13, 21, 103, 173. Several studies indicate that excitatory synapses exhibit the most important alterations in HD 47, 79, 160, and numerous reports have concluded that corticostriatal and hippocampal synaptic plasticity are impaired (Table 2). These studies suggest that synaptic deficits, including alterations in synaptic transmission, plasticity and aberrant spine density/morphology, may be the initial triggers of the cognitive deficits observed in HD 204. Interestingly, a recent study using a corticostriatal co‐culture cell model from wild‐type and YAC128 mice demonstrated impaired development of excitatory synaptic connectivity and reduced dendritic complexity in YAC128 MSNs. Moreover, a number of other HD‐associated synaptopathy features previously reported in the striatum of HD mice at mid‐to‐late stages 245 were also observed 37. Remarkably, mutant huntingtin is required pre‐ and post‐synaptically to elicit these effects, which is in agreement with previous findings showing that its expression selectively in either striatal or cortical neurons is insufficient to fully recapitulate HD behavioral and neuropathological phenotype 110, 111, 323. In the same line, the absence of mutant huntingtin in cortical afferents only partially improves striatal neuronal activity and behavior in a conditional mouse model of HD 76. Altogether, these studies point to the existence of cell–cell pathogenic interactions that shape the progression of striatal deficits.
ALTERED MECHANISMS UNDERLYING SYNAPTIC DYSFUNCTION AND COGNITIVE IMPAIRMENT IN HD
Neuronal dysfunction and cognitive impairment precede motor symptoms and neuronal death in HD patients, and they occur long before (or in the absence) of cell death in HD mouse models, suggesting that the early cognitive deficits may be a consequence of synaptic dysfunction rather than of cell loss 180, 194, 210, 269, 316. In fact, huntingtin binds to a wide range of intracellular proteins, many of them responsible for synaptic transmission 275, and mutant huntingtin is present in pre‐synaptic 216, 281 and post‐synaptic terminals 294, where it interacts with several synapse‐related proteins impairing synaptic function in HD mice 147, 281, 354. Moreover, the presence of mutant huntingtin alters numerous cellular and molecular mechanisms including protein aggregation, protein–protein interaction, calcium signaling, mitochondrial function, transcriptional regulation and chromatin remodeling, vesicle transport, neurotransmitter release and receptor activity 294, 351, 354. Potentially, all these alterations can impact on neuronal functioning, synaptic plasticity and, ultimately, cognitive function. Nevertheless, in the following sections we will mainly focus on those altered mechanisms that have been demonstrated to participate in neuronal dysfunction leading to synaptic plasticity deficits and/or cognitive impairment.
Spine loss
Dendritic spines play critical roles in synaptic transmission and plasticity because changes in their morphology and density modulate the formation and maintenance of the synapses, enabling the dynamics of neural circuitry 274, 309, 320. Several studies demonstrate synaptic alterations in MSNs neurons 81, 106 and in prefrontal cortical pyramidal neurons 283 from HD post‐mortem brain samples. Furthermore, studies in HD mouse models also provide evidence for altered dendritic morphology (Table 2). Here, we describe the mechanisms that contribute to these alterations.
RhoGEF/GAP signaling: Kalirin‐7
Kalirin‐7, a postsynaptic brain‐specific guanine exchange factor protein for Rho‐like small GTPases 223 has emerged as a key regulator of excitatory synapses. Overexpression of Kalirin‐7 causes an increase in dendritic spine density, spine size and synapse number, while Kalirin‐7 knockdown promotes spine shrinkage and loss in cultured hippocampal and cortical neurons 174, 175, 221, 222. Importantly, and consistent with in vitro experiments, these alterations in excitatory synapses correlate with a decline in the magnitude of hippocampal LTP, reduced glutamatergic synaptic transmission in the cortex, and impaired cognitive function 39, 175, 221, 222, 335, thus supporting a role for Kalirin‐7 in learning and memory processes through the modulation of structural plasticity. Interestingly, Kalirin interacts with huntingtin‐associated protein 1 55, and alterations in excitatory synapses occur at early disease stages in HD mouse models 47, 79, 172. These synaptic alterations have been suggested as contributing to cognitive symptoms in both HD patients and animal models 166, 197, 304, 313. In a recent study, and following a broad analysis of synaptic‐related proteins such as NMDA (GluN1 and GluN2B) and AMPA (GluA1) receptor subunits, presynaptic (VGlut1 and synaptophysin) and postsynaptic (Kalirin‐7, PSD95, Shank3 and CaMKII) scaffolding and signaling proteins, only Kalirin‐7 showed an early and specific reduction in the cortex of HdhQ7/Q111 and R6/1 mice, which correlates with cortical dendritic spine alteration, impaired corticostriatal synaptic plasticity and motor and procedural learning behavioral deficits in 2‐ and 6‐month‐old HdhQ7/Q111 mice 239. Supporting the hypothesis that loss of Kalirin‐7 in the cortex of young HD mice could be associated with the early loss of excitatory synapses in HD, Kalirin‐7 overexpression restores the number of cortical glutamatergic synapses in mature cultured HD cortical neurons 239.
GluN3A
NMDARs play crucial roles in remodeling and maintaining excitatory synapses, and their activity is altered in MSNs from HD mice 272. NMDAR hyper‐function can be detected in HD MSNs at early stages 159, 193, well before synapse and spine loss, behavioral deficits and neuronal death, pointing to signaling through these receptors as a key player in the HD pathogenic cascade.
PACSIN1 controls the endocytic removal of GluN3A‐containing NMDARs 224. GluN3A is highly expressed in the brain during early postnatal development to prevent premature synapse plasticity and stabilization, but its expression declines afterwards 135, 179, 247, 328. Nevertheless, a recent study found that mutant huntingtin binds to and sequesters PACSIN1, causing its subcellular redistribution away from the synapse and promoting accumulation of GluN3A‐containing NMDARs at the surface of striatal neurons. In agreement, GluN3A levels are increased in human HD striatum and in striatal membrane fractions obtained from distinct HD mouse models including R6/1, YAC128 and HdhQ111 knock‐in mice 179, suggesting that this redistribution of GluN3A has a pathological role. The contribution of GluN3A reactivation in MSNs as an important factor in HD pathogenesis receives support from the finding that overexpression of GluN3A replicates the reduced synaptic connectivity observed in MSNs from YAC128 mice, whereas lack of GluN3A corrects the early enhancement of NMDAR currents and prevents both early and progressive dendritic spine pathology in MSNs from YAC128 mice. Importantly, it also ameliorates striatum‐dependent motor and cognitive decline 179. Thus, these studies reveal a novel early disease mechanism that mediates NMDAR dysfunction and synapse loss in HD MSNs.
Store‐operated Ca2+ entry
GluN3A deletion experiments suggest that GluN3A expression is necessary to/permissive of HD pathogenesis 179. Nevertheless, other parallel mechanisms seem to be set in motion by the presence of mutant huntingtin, leading to spine loss in HD MSNs. Dysregulation of intracellular neuronal Ca2+ signaling plays a role in HD progression 17, 18, 191. Mutant huntingtin interacts with type 1 inositol‐1,4,5‐trisphosphate receptor (InsP3R1), a neuronal endoplasmic reticulum (ER) Ca2+ release channel, causing its overactivation and excessive Ca2+ release from the ER 295, 296. Ca2+ release from the ER stimulates neuronal store‐operated Ca2+ entry (nSOC) channels in the plasma membrane 240, and this pathway plays an important role in the maintenance of postsynaptic mushroom spines in hippocampal neurons 233, 293, 347. Interestingly, elevated nSOC was reported in cultured MSNs from YAC128 mice 332, suggesting that aberrant InsP3R1 function and disrupted ER Ca2+ homeostasis could contribute to spine loss in MSNs in HD. Interestingly, InsP3R1 knockdown suppresses Ca2+ leak from the ER, reduces nSOC levels in spines and is sufficient to prevent spine loss in YAC128 MSNs 331.
Impaired neurotrophic signaling: TrkB/p75NTR imbalance
A deficit in neurotrophic support is considered a key player in HD neuropathology. In particular, a reduction in BDNF protein levels was reported in several brain regions of HD patients and mouse models 83, 96, 348, 349, 350, 351. However, this general finding has not been replicated in a recent study where cortical BDNF mRNA levels were assessed in BACHD and heterozygous Q175 knock‐in mice using multiple primers and reference genes 227, thus challenging the view that this neurotrophic deficit is a major pathogenic mechanism in HD.
BDNF exerts trophic effects by binding to its receptors TrkB and p75NTR. BDNF binding to TrkB receptor has been shown to promote neuronal survival 207, 279, while BDNF binding to p75NTR could either potentiate Trk function 20, 42, 176, 282 or activate cell death cascades 56, 86, 303. Notably, whereas BDNF induces hippocampal LTP through TrkB 230, p75NTR has been involved in regulating LTD without affecting LTP 251, 329 [for review see Lu et al. 169]. These alterations in synaptic transmission are associated with structural changes. Thus, a deficiency in BDNF‐mediated intracellular signaling causes dendritic abnormalities in the striatum and cerebral cortex 8, 105, while activation of p75NTR blocks axonal and dendritic elongation and arborization by activation of RhoA, a Rho GTPase that negatively regulates neurite elongation and actin assembly 329, 341.
Importantly, reduced TrkB levels have been reported in HD cellular and mouse models, as well as in HD patients 93, 95, 352. Moreover, there is increased p75NTR mRNA expression in the caudate, but not in the cortex, of HD patients 352. Therefore, there is an imbalance between TrkB and p75NTR expression in the caudate nucleus of HD patients, and in the striatum and hippocampus, but not in the cortex, of HD mouse models at early stages of the disease 28, 29, suggesting that this imbalance contributes to early and progressive HD pathology. Interestingly, genetic normalization of p75NTR in HdhQ7/Q111 mice rescues hippocampal synaptic plasticity and memory function, and prevents hippocampal dendritic spine alterations, likely by normalization of RhoA GTPase activity 28, 190. Reinforcing the role of p75NTR in cognitive deficits in HD, overexpression of p75NTR in the hippocampus of wild‐type animals reproduces those memory deficits observed in HD mice 28, while specific hippocampal p75NTR knockdown prevents the manifestation of cognitive impairment. Together, these findings demonstrate that p75NTR upregulation plays a role in the synaptic and learning and memory deficits observed in HD mice. In agreement with these data, overexpression of p75NTR in hippocampal neurons decreases spine density 345, while null p75NTR mice exhibit increased hippocampal dendritic spine density, improved spatial learning and enhanced LTP 10, 108. In addition, Plotkin et al. have shown that although cortical production of BDNF, its delivery to the striatum and activation of TrkB are normal in BACHD and heterozygous knock‐in zQ175 HD mice, BDNF fails to support corticostriatal LTP specifically at corticostriatal synapses of the indirect pathway. Importantly, this kind of plasticity can be rescued by knocking down p75NTR or inhibiting its downstream targets RhoA, ROCK and phosphatase and tensin homolog deleted on chromosome 10 (PTEN), indicating that enhanced signaling through p75NTR and PTEN antagonizes TrkB function and corticostriatal LTP 227. It remains to be determined whether this improvement in corticostriatal connectivity can be reproduced in other HD models, and whether it translates into an amelioration of corticostriatal‐dependent learning and/or motor symptoms.
COGNITIVE IMPAIRMENT
Adenosine type 2A receptor
HD mouse models display reduced reversal learning and working memory deficits 30, 31, 32, 33, 34, 35, 36, 166, 183, 304, 313 like early HD patients 100, 214. This type of cognitive impairment, which significantly affects the patient's quality of life, represents a primary dysfunction of the corticostriatal pathway 44, 47. In a recent study, Chen and coworkers 164 evaluated the ability of adenosine type 2A receptor (A2AR) inactivation to reverse the deficits in working memory and synaptic plasticity at early stages of HD. Their results show that genetic or pharmacological inactivation of A2AR prevents working memory deficits in R6/2 mice, without modifying motor dysfunction. Moreover, although wild‐type and R6/2 mice display similar LTD and LTP at corticostriatal synapses, and pharmacological blockade of A2AR inhibits LTP to a similar extent in both genotypes, it selectively reduces LTD amplitude in mutant mice 164. Thus, striatal A2AR emerges as a novel target to fight against the cognitive inflexibility, namely working memory impairment, reported in the prodromal phase of HD 214, 291.
PKA overactivation
Synaptic plasticity and learning and memory processes depend on an appropriate balance between kinase and phosphatase activities 137, 178, 182. Changes in the expression and activity of different phosphatases 73, 80, 189, 226, 258, 260, 262, 333, 337, 338 and kinases 5, 78, 94, 102, 212, 256, 257, 260, 262, 336 (and reviewed by Bowles and Jones 27) have been reported in HD models and human brain, suggesting that aberrant function of these proteins likely contributes to HD pathogenesis. Although alterations in distinct phosphatases can potentially contribute, directly or indirectly, to synaptic plasticity deficits and cognitive decline in HD, this issue remains to be directly addressed 100. Conversely, it has been demonstrated that adenosine 3′5′ cyclic‐monophosphate (cAMP)‐dependent protein kinase (PKA) overactivation contributes to hippocampal‐dependent synaptic and cognitive deficits in exon‐1 HD mice 102, 306.
The PKA pathway is known to regulate specific types of long‐term synaptic plasticity (reviewed by Nguyen and Woo 202), and to play a critical role in hippocampal‐dependent learning and memory formation 1, 132, but persistent and aberrant activation of PKA can lead to memory impairment 1, 25, 71, 136, 226. Likewise, increased hippocampal PKA activity leads to cognitive dysfunction in R6/2 mice as demonstrated by the beneficial effect of intra‐hippocampal injection of Rp‐cAMPs, a PKA inhibitor, on recognition memory 102. Reinforcing these findings, cAMP levels are higher in hippocampal nerve terminals from R6/1 mice than in controls, and dopamine type 1 (D1) and A2A receptors display increased response to their ligands in mutant mice. This leads to PKA overactivation in the hippocampus and participates in an occlusion mechanism 306. In fact, a combined chronic blockade of D1R and A2AR, but not a single acute or a chronic blockade of either receptor alone, normalizes PKA activity in the hippocampus and ameliorates cognitive dysfunction in R6/1 mice 306. Moreover, and in contrast to vehicle‐injected mutant mice, R6/1 animals injected daily with SCH23390 (D1R antagonist) plus SCH58261 (A2AR antagonist) display a significant LTP induction in vivo. Overall, these data show that D1R and A2AR blockade normalizes hippocampal PKA activity, enhances synaptic potentiation at the CA3‐CA1 region and ameliorates cognitive dysfunction in R6/1 mice 306.
In HD, non‐motor symptoms include sleep and circadian disturbances (reviewed by Morton 198). Interestingly, pathological up‐regulation of cAMP/PKA signaling has been implicated in sleep and activity abnormalities in fly HD models. Elevated PKA activity in healthy flies produces patterns of sleep and activity similar to those found in flies expressing mutant huntingtin, whereas genetic reduction of PKA abolishes sleep/activity deficits, restores the homeostatic response and extends lifespan in HD flies. Remarkably, decreasing PKA also prevents immediate memory impairment in HD model flies 104.
Summing up, aberrant PKA activity may be a general consequence of mutant huntingtin expression, and may underlie neuronal dysfunction in distinct brain areas and several HD phenotypes. For instance, abnormalities in sleep and circadian rhythms have a negative impact on cognitive, emotional and psychiatric function.
Deficient cGMP signaling
The nitric oxide/soluble guanylyl cyclase/3′,5′‐cyclic guanosine monophosphate/cGMP‐dependent protein kinase (NO/sGC/cGMP/cGK) signaling pathway has been widely implicated in synaptic plasticity, and in learning and memory in several brain regions, including the hippocampus (reviewed by Kleppisch and Feil 143). Interestingly, neuronal NOS (nNOS) mRNA levels are decreased in the caudate of HD patients 205, and changes in nNOS protein levels also occur in the striatum and cortex of HD mouse models 63, 64, 65, 125, 225. The study of the integrity of the nNOS/cGMP pathway in the hippocampus of HD mice, and of its potential contribution to hippocampal learning and memory deficits, has shown that both nNOS and cGMP levels are significantly reduced in the hippocampus of R6/1 mice, and that an acute post‐training injection with sildenafil, a selective inhibitor of the cGMP‐specific phosphodiesterase (PDE) 5 24, increases cGMP levels and improves novel object recognition memory and passive avoidance learning 261. These data support the idea that decreased hippocampal cGMP levels contribute to cognitive dysfunction in R6/1 mice. Another study performed in the rat 3‐nitropropionic acid toxic model of HD shows that sildenafil treatment improves memory performance in the Morris water maze 298. Importantly, cGMP levels are also reduced in the hippocampus of HD patients 261. Thus, PDE5 inhibition may prove to be beneficial in ameliorating hippocampal‐dependent cognitive deficits in HD.
Transcriptional dysregulation: CREB and its co‐activator CBP
Transcriptional dysregulation has been shown in HD human brain, as well as in in vivo and in vitro disease models 67, 120, 155, 170, 171, 181, 258. Mutant huntingtin can cause transcriptional dysregulation by (i) sequestration of positive transcriptional regulators such as TATA‐binding protein 267, specificity protein‐1 163 or cAMP‐response element binding protein (CREB) binding protein (CBP) 267, 289; (ii) loss of interaction with negative transcriptional regulators, such as the repressor element‐1 transcription/neuron restrictive silencer factor (NRSE), resulting in REST/NRSF complex nuclear translocation and transcriptional repression of several neuronal‐specific genes 353 and (iii) increasing ubiquitination and histone methylation, and reducing histone acetylation 273.
CREB is a transcription factor that mediates stimulus‐dependent changes in the expression of genes critical for neuronal survival, plasticity and growth 152, 154, 167, 168. CREB activity is regulated by phosphorylation at serine 133 (Ser133) and at additional sites, as well as by association with CREB co‐activators 253. Indeed, CREB phosphorylation at Ser133 facilitates the binding of the transcriptional co‐activator CBP 53, 146, 215, 242. The interaction between CREB and CBP, or other members of the transcriptional machinery, facilitates gene expression 84, 215. In fact, CBP has emerged as a key regulator of CREB‐mediated transcription by acting as a CREB transcriptional co‐activator 26, 53, 244 and as a histone acetyltransferase (HAT) to disrupt repressive chromatin structure and allow gene transcription 48, 130, 208, 318. Expression of CREB‐related target genes is downregulated in several in vitro and in vivo models of HD 96, 206, 292. Moreover, mutant huntingtin interacts with CBP and blocks its HAT activity 288. Importantly, hypoacetylation of histone H3 is associated with downregulation of genes in R6/2 mice and knock‐in cell lines 263.
Activity‐induced gene transcription is required for normal hippocampal synaptic plasticity and memory consolidation 9, 131, and compelling evidence indicates that CREB is essential for activity‐induced memory gene expression 15, 276. Additionally, several studies show reduced chromatin acetylation and hippocampal LTP and long‐term memory (LTM) deficits in mouse models with compromised CBP activity 2, 51, 209, 266, 308, 330. In line with these data, CBP levels are reduced in the hippocampus of HD patients and HdhQ7/Q111 mice in close correlation with the presence of spatial and recognition memory deficits 98. Moreover, reduced CBP levels in HdhQ7/Q111 mice are associated with selective dysregulation of CREB/CBP‐target genes related to memory and synaptic plasticity (c‐fos, Nr4a2 and Arc) 98. Reduced CBP expression and/or activity have been associated with diminished H3 acetylation in mouse models of cognitive dysfunction 51, 144, 255. Consistently, decreased hippocampal CBP levels are paralleled by diminished H3 acetylation in HdhQ7/Q111 mice, suggesting that lower CBP levels and decreased histone acetylation are responsible, at least in part, for memory dysfunction in HD 98. In agreement with studies showing that CBP HAT activity plays a crucial role in memory consolidation processes 144, and that trichostatin A (TSA), a general histone deacetylase inhibitor, enhances hippocampal‐dependent memory consolidation by increasing the expression of specific genes during memory consolidation 315, TSA reverses LTM impairment in HdhQ7/Q111 mice, accompanied by increased levels of c‐fos and Arc 98.
THERAPEUTIC STRATEGIES
Currently, there is no known effective treatment for cognitive dysfunction in HD as so far clinical trials have tested traditional cognitive enhancers and anti‐depressants without signs of efficacy [reviewed by Killoran and Biglan 139].
The fact that in HD neuronal death does not occur until late stages of the disease suggests that neuronal dysfunction and abnormal synaptic plasticity occur earlier and are responsible for cognitive decline, which opens a window for therapeutic interventions. Moreover, targeting those early pathophysiological events is likely to provide better therapeutic outcomes than trying to prevent cell death once neurons are severely affected. As explained above, the presence of mutant huntingtin alters distinct cellular and molecular mechanisms, all of which can directly or indirectly impact on neuronal functioning, leading to synaptic and cognitive impairment. In this line, several genetic, pharmacological and non‐pharmacological approaches have proved beneficial in HD models, not necessarily because they specifically target an altered mechanism or signaling cascade that participates in neuronal dysfunction leading to cognitive impairment, but because they improve the expression of receptor subunits, neurotrophic factors or other molecules involved in plasticity processes, and/or because they reduce the level of detrimental effectors. Next, we provide one such example. PDEs are the enzymes responsible for cAMP/cGMP degradation that through their different subcellular distribution allow compartmentalization and tight temporal and spatial control of cyclic nucleotide signaling 14. PDE inhibitors are increasingly being considered as cognitive enhancers 22, 241, 278, 339, and the cognitive‐enhancing properties of a PDE10A inhibitor were recently examined in R6/1 mice. PDE10A, a cAMP/cGMP dual‐substrate PDE, is enriched in nuclear fractions both in wild‐type and R6/1 mouse hippocampus, without differences in its levels or intracellular distribution between genotypes. Chronic treatment with papaverine, a PDE10A inhibitor, improves spatial and object recognition memory in R6/1 mice, and likely works through the activation of the PKA pathway, as the phosphorylation level of distinct cGK substrates is not modified in either genotype 101. These results appear to contradict the finding that increased PKA signaling in R6 mice hippocampus leads to hyperphosphorylation of PKA membrane targets implicated in synaptic plasticity and learning and memory, and to impairment of object recognition and spatial memory 102, while targeting aberrant PKA signaling improves hippocampal‐dependent synaptic and cognitive function 102, 306. Overall, these studies support the idea that hyperactivity of hippocampal PKA in HD mice is not a global event, but rather restricted to specific subcellular domains 101, 102. In fact, the memory‐enhancing effect of papaverine in R6/1 mice likely involves a partial, but significant, recovery of GluA1 phosphorylation levels together with increased CREB phosphorylation in the hippocampus 101. Thus, PDE10 might be a good therapeutic target to improve hippocampal cognitive impairment in HD. Remarkably, although striatal PDE10A levels are reduced well before motor symptom onset in HD 203, as confirmed by a study with the radioligand [18F] MNI‐659A 259, PDE10 is being targeted for striatal dysfunction. Indeed, PDE10A inhibitors have attracted interest as potential novel pharmacotherapies for HD 91, 92, 114, 116, 142, with ongoing clinical trials 116. Nevertheless, in this section we chose to focus on those strategies with therapeutic potential for treating cognitive dysfunction in HD whose choice is justified by the identification of an affected pathway.
Functional and morphological changes in the neocortex have been proposed as initial triggers of striatal pathology in HD. In this context, there is an early and specific reduction in cortical Kalirin‐7 levels in HD mice, paralleled by early cortical dendritic spine alteration, impaired corticostriatal LTP and cognitive deficits 239. It is noteworthy that the number of cortical glutamatergic synapses in cultured HD neurons can be restored upon Kalirin‐7 overexpression 239. Although the study of the impact of Kalirin‐7 overexpression in adult HD mice is hampered by methodological limitations, the early loss of Kalirin‐7 could contribute not only to decreased spine density, but probably also to altered corticostriatal synaptic transmission and cognitive deficits. In summary, the identification of Kalirin‐7 downregulation at early HD stages and its role in modulating HD cortical excitatory synapses 239 allows us to hypothesize that cortical function could be restored by increasing the levels of Kalirin‐7. Such increase could be the first step to prevent subsequent loss of corticostriatal connectivity, striatal dysfunction and, later, striatal neuronal degeneration.
Concerning spine loss in MSNs, two mechanisms have been elucidated. One involves aberrant reactivation of juvenile GluN3A subunits, which promotes early and progressive dendritic spine pathology that likely underlies cognitive and motor impairment, and ultimately, neuronal death. In fact, lack of GluN3A ameliorates striatum‐dependent cognitive and motor decline, and reduces striatal cell death in YAC128 mice 179, which has led to the proposal that GluN3A might be a good target for therapeutic approaches in HD 92. The other mechanism is associated with dysregulation of intracellular neuronal Ca2+ signaling, as enhanced nSOC has been shown to cause synaptic loss in HD MSNs 331. Interestingly, inhibition of nSOC with EVP4593 not only prevents spine loss in YAC128 MSNs, in vitro and in vivo 331, but also protects cultured HD MSNs against glutamate toxicity and improves motor symptoms in a fly model of HD 332, thus supporting the possibility that targeting nSOC could have a beneficial impact on HD.
Another crucial regulator of synaptic plasticity and neuronal survival proposed as an excellent therapeutic target for treating the clinical hallmarks of HD is BDNF 3, 351, 354. However, BDNF administration has shown important methodological drawbacks in HD models 349, and several studies indicate that increasing BDNF levels only partially improves HD phenotype 6, 89, 97, 277, 334. Previous studies conducted by our group and others demonstrate reduced TrkB levels in patients and in different HD models 93, 95, 352. Together with the emerging evidence of imbalanced TrkB and p75NTR expression/signaling in HD 28, 29, 221, 227, 352, this could contribute to the incomplete reversion of HD pathology by the administration of the neurotrophin.
Although a general genetic reduction of p75NTR levels in HdhQ7/Q111 mouse brain does not prevent motor learning deficits or corticostriatal LTP abnormalities, the levels of DARPP‐32, a striatal marker known to be reduced in HD mice from early stages 19, are reestablished 28. These results suggest that striatal neuronal dysfunction can be slightly ameliorated, but not prevented, by downregulation of aberrant p75NTR expression in HdhQ7/Q111 brain. On the other hand, specific knockdown of striatal p75NTR reverses corticostriatal LTP abnormalities in BACHD mice 227. In this context, it is important to note that p75NTR levels are not altered in the cerebral cortex of HD mice and patients 28, 29, suggesting that decreased cortical p75NTR levels in HD mice can be deleterious for synaptic plasticity and cognitive processes. Altered expression of TrkB and p75NTR receptors disturbs BDNF‐induced neuronal protection in a cellular HD model, and reduction of p75NTR levels in corticostriatal slices of HdhQ111/Q111 mice not only increases cell survival, but also prevents the cell death induced by BDNF 29. Therefore, targeting p75NTR has the potential to improve corticostriatal plasticity and reduce cell death in HD.
Besides impairment in corticostriatal connectivity, disturbances in hippocampal function contribute to HD memory deficits in middle disease stages. Thus, hippocampal dysfunction is an important hallmark of HD pathology, and preservation/restoration of hippocampal function could represent a promising alternative strategy to prevent memory loss in HD. In line with recent studies supporting a critical role for p75NTR in hippocampal‐dependent synaptic plasticity 10, 108, normalization of hippocampal p75NTR levels in distinct HD mouse models with genetic or pharmacological approaches rescues hippocampal synaptic plasticity, memory deficits and dendritic spine alterations, likely by normalization of the RhoA GTPase activity 28, 190. Overall, this evidence suggests that antagonism of p75NTR could represent an excellent approach to promote BDNF‐mediated signaling in HD corticostriatal pathway, thereby restoring corticostriatal connectivity 227, ameliorating hippocampal synaptic dysfunction and memory deficits 135 and improving cell survival 29. An important consequence of these findings is that whereas TrkB is widely and robustly expressed in the adult brain, p75NTR has a restricted tissue distribution, and its expression is developmentally downregulated in most parts of the brain, which makes the targeting of p75NTR likely to have fewer side effects in HD patients. Remarkably, chronic administration of fingolimod (FTY720), an immunomodulatory drug used in the treatment of multiple sclerosis patients 52, ameliorates LTM deficits and dendritic spine loss in CA1 hippocampal neurons from R6/1 mice, and these effects are accompanied by normalization of p75NTR levels and reduced astrogliosis in the hippocampus 190. Interestingly, chronic administration of FTY720 improves motor function, prolongs survival and reduces brain atrophy in R6/2 mice, and these effects are accompanied by increased BDNF levels, strengthening of neuronal activity and connectivity, reduction of mutant huntingtin aggregates and increased phosphorylation of mutant huntingtin in residues predicted to attenuate its toxicity 68. It would be useful to evaluate whether the normalization of p75NTR levels and reduction of astrogliosis reported in the hippocampus of FTY720‐treated R6/1 mice 190 also occur in the striatum.
Aberrant PKA signaling promotes hippocampal‐dependent synaptic and memory impairment in HD mice 102, 306. Notably, abnormal PKA signaling is also responsible for sleep disturbances in fly HD models, leading to the proposal of sleep and cAMP/PKA as prodromal indicators of disease, and therapeutic targets for intervention 104. Increased signaling through D1R and A2AR contributes to PKA overactivation and hippocampal‐dependent synaptic and memory impairment in HD mice 306. Based on the finding that PKA activity is also increased in the hippocampus of HD patients 102, we propose that targeting D1R and A2AR might be a therapeutic approach to improve hippocampal‐dependent cognitive function in HD. The combined antagonism of the two receptors normalizes striatal PKA activity, but does not ameliorate motor deficits 306, and inactivation of A2AR prevents working memory deficits in R6/2 mice, but does not modify motor dysfunction either 137. Actually, although striatal A2AR emerges as a novel target to fight against cognitive inflexibility, namely working memory impairment 164, this exciting therapeutic possibility needs to be carefully considered since targeting A2AR for HD motor symptoms remains largely controversial. Some studies demonstrate a neuroprotective effect of an A2AR antagonist in toxic models of HD, while others report no recovery from motor deficits or a delayed deterioration of motor performance after treatment with A2AR agonists in R6/2 mice [reviewed by Lee and Chern 153, Popoli et al. 231 and Popoli 232]. Thus, targeting A2AR in HD is a puzzling issue, and the therapeutic window for A2AR antagonists might be restricted to the early phases of HD. Conversely, activation of A2AR normalizes synaptic activity in the striatum of symptomatic R6/2 mice, and may thus help to restore corticostriatal connectivity at later stages of the disease 44.
In contrast to cAMP levels 306, hippocampal cGMP levels are reduced in HD mice 261. Since targeting the cGMP‐specific PDE5 improves cGMP levels and ameliorates hippocampal‐dependent learning and memory in R6/1 mice, and cGMP levels are also diminished in the hippocampus of HD patients 261, normalization of cGMP levels emerges as an approach to counteract deficits in hippocampal cognitive function in HD. Given that the nNOS pathway is highly affected in the striatum and cortex too 63, 64, 65, 125, 225, it is tempting to speculate that targeting this pathway might also ameliorate corticostriatal dysfunction. So far, two studies have shown that the PDE5 inhibitor sildenafil protects against biochemical and behavioral abnormalities in the 3‐nitropropionic acid toxic model of HD 238, 298, but whether sildenafil or other PDE5 inhibitors improve corticostriatal connectivity, corticostriatal‐dependent learning and/or motor dysfunction in genetic models of the disease remains to be addressed. It is noteworthy that cGMP can promote mitochondrial biogenesis and ATP synthesis 195, which is relevant because mitochondrial function is known to be compromised in HD 50. Furthermore, PDE5 inhibitors have emerged as a potential therapeutic strategy to improve not only cognitive function, but also to target neuroinflammation and neurodegeneration (reviewed by Peixoto et al. 220), and thus they could be valuable multi‐purpose drugs in the context of HD. Moreover, it is expected that the use of PDE5 inhibitors will prove to be of therapeutic interest because under physiological conditions transient elevations in striatal intracellular cGMP levels increase neuronal excitability and facilitate spontaneous and evoked corticostriatal transmission 302, which would improve connectivity in HD. Interestingly, intrastriatal infusion or systemic administration of the selective PDE10A inhibitor TP‐10 increases the responsiveness of striatal MSNs to cortical input 301, and this effect depends on the NO‐sGC‐cGMP signaling cascade 211. Considering these findings and the reports on reduced nNOS mRNA in postmortem tissue from HD subjects 205 and in HD mouse models 64, 225, it is likely that a combination of both PDE10A inhibitors and sGC activators will be useful to improve corticostriatal transmission in HD.
As an alternative, or complementary, strategy to restore hippocampal function in HD at middle and advanced disease stages, we propose the modulation of CBP levels and/or activity. CBP levels are reduced in the hippocampus of HdhQ7/Q111 mice, where they are accompanied by diminished histone 3 acetylation and spatial and recognition memory deficits 98. Since treatment with a general HDAC inhibitor reverses LTM impairment in HdhQ7/Q111 mice, CBP loss of function may result in decreased memory‐related gene transcription and be responsible, at least in part, for the spatial and recognition memory deficits observed in HdhQ7/Q111 mice 98. Remarkably, several studies suggest an important role of CBP loss of function in polyQ‐dependent striatal neurodegeneration in HD models 64, 128, 185, 206, 297. It is noteworthy that restoration of CBP striatal function by overexpression of CBP or by using HDAC inhibitors improves striatal atrophy and survival, as well as motor symptoms in HD models 82, 88. Therefore, therapies aimed at increasing CBP levels and/or activity by using HDAC inhibitors hold the promise to be a good approach to prevent corticostriatal‐ and hippocampal‐dependent dysfunction, motor symptoms and, ultimately, neurodegeneration. Remarkably, recent studies have found that HDACi 4b, a HDAC1/3 inhibitor, has beneficial transgenerational effects in HD mice through altered DNA and histone methylation 126, while RGFP966, an inhibitor of HDAC3, improves motor deficits on rotarod and open field, accompanied by neuroprotective effects on striatal volume and decreased glial fibrillary acidic protein immunoreactivity in the striatum of N171‐82Q mice 127. Therapies targeting transcriptional dysregulation in HD include sodium phenylbutyrate (phase I) and HDACi 4b (preclinical) 145. Even though research in this area is still at a preliminary stage, and crucial issues need to be addressed, such as the development of new potent and more selective HDAC inhibitors, with excellent blood–brain‐barrier permeability, less cytotoxicity and reduced side effects, HDAC inhibitors show promise as a new avenue for therapeutic interventions in HD.
CONCLUDING REMARKS
In summary, we have reviewed new evidence for early cortical and corticostriatal dysfunction in HD followed by hippocampal dysfunction, prior to the manifestation of motor symptoms, driving the search for novel therapeutic approaches to improve HD pathology at different disease stages. In particular, we propose that a first therapeutic intervention has to be focused on preserving/restoring corticostriatal connectivity, which would impact on intrinsic striatal function. In this context, given that reduced levels of Kalirin‐7 are responsible, at least in part, for the early altered corticostriatal function, we propose the preservation/restoration of Kalirin‐7 levels as an early therapeutic intervention to maintain functional cortical excitatory synapses. In the case of MSNs, recent evidence indicates that spine loss in YAC128 neurons is associated with reactivation of juvenile GluN3A. Although the downstream events are not fully characterized, these findings support the development of GluN3A‐selective antagonists and/or alternative therapeutic approaches to block abnormal GluN3A expression. Aberrant InsP3R1 activity leading to reduced ER Ca2+ levels and increased spine SOC are also implicated in MSNs spine loss, suggesting that targeting nSOC in MSNs might prove to be useful. Targeting the PKA pathway may ameliorate hippocampal plasticity and cognitive function in HD. To improve not only corticostriatal dysfunction, but also hippocampal‐dependent deficits in HD, we propose (i) genetic or pharmacological inhibition of p75NTR to preserve synaptic plasticity and cognitive function, as well as prevent striatal neuronal death, and (ii) treatment with HDAC inhibitors that exhibit promising therapeutic effects in restoring memory and improving striatal survival and motor coordination at more advanced disease stages. Data obtained so far indicate that inhibition of PDE5 ameliorates hippocampal‐dependent learning and memory, but further studies are needed to address the therapeutic potential of targeting cGMP signaling using PDE inhibitors to improve corticostriatal dysfunction in HD. The use of PDE10A inhibitors will likely prove to be beneficial by improving both hippocampal and corticostriatal deficits in HD.
REFERENCES
- 1. Abel T, Nguyen PV (2008) Regulation of hippocampus‐dependent memory by cyclic AMP‐dependent protein kinase. Prog Brain Res 169:97–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 2. Alarcon JM, Malleret G, Touzani K, Vronskaya S, Ishii S, Kandel ER, Barco A (2004) Chromatin acetylation, memory, and LTP are impaired in CBP+/− mice: a model for the cognitive deficit in Rubinstein‐Taybi syndrome and its amelioration. Neuron 42:947–959. [DOI] [PubMed] [Google Scholar]
 - 3. Alberch J, Perez‐Navarro E, Canals JM (2004) Neurotrophic factors in Huntington's disease. Prog Brain Res 146:195–229. [DOI] [PubMed] [Google Scholar]
 - 4. Andre VM, Cepeda C, Venegas A, Gomez Y, Levine MS (2006) Altered cortical glutamate receptor function in the R6/2 model of Huntington's disease. J Neurophysiol 95:2108–2119. [DOI] [PubMed] [Google Scholar]
 - 5. Apostol BL, Illes K, Pallos J, Bodai L, Wu J, Strand A et al (2006) Mutant huntingtin alters MAPK signaling pathways in PC12 and striatal cells: ERK1/2 protects against mutant huntingtin‐associated toxicity. Hum Mol Genet 15:273–285. [DOI] [PubMed] [Google Scholar]
 - 6. Arregui L, Benitez JA, Razgado LF, Vergara P, Segovia J (2011) Adenoviral astrocyte‐specific expression of BDNF in the striata of mice transgenic for Huntington's disease delays the onset of the motor phenotype. Cell Mol Neurobiol 31:1229–1243. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 7. Backman L, Robins‐Wahlin TB, Lundin A, Ginovart N, Farde L (1997) Cognitive deficits in Huntington's disease are predicted by dopaminergic PET markers and brain volumes. Brain 120:2207–2217. [DOI] [PubMed] [Google Scholar]
 - 8. Baquet ZC, Gorski JA, Jones KR (2004) Early striatal dendrite deficits followed by neuron loss with advanced age in the absence of anterograde cortical brain‐derived neurotrophic factor. J Neurosci 24:4250–4258. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 9. Barco A, Pittenger C, Kandel ER (2003) CREB, memory enhancement and the treatment of memory disorders: promises, pitfalls and prospects. Expert Opin Ther Targets 7:101–114. [DOI] [PubMed] [Google Scholar]
 - 10. Barrett GL, Reid CA, Tsafoulis C, Zhu W, Williams DA, Paolini AG et al (2010) Enhanced spatial memory and hippocampal long‐term potentiation in p75 neurotrophin receptor knockout mice. Hippocampus 20:145–152. [DOI] [PubMed] [Google Scholar]
 - 11. Bates GP, Mangiarini L, Mahal A, Davies SW (1997) Transgenic models of Huntington's disease. Hum Mol Genet 6:1633–1637. [DOI] [PubMed] [Google Scholar]
 - 12. Begeti F, Schwab LC, Mason SL, Barker RA. Hippocampal dysfunction defines disease onset in Huntington's disease. J Neurol Neurosurg Psychiatry 87:975–981. [DOI] [PubMed] [Google Scholar]
 - 13. Behr J, Wozny C, Fidzinski P, Schmitz D (2009) Synaptic plasticity in the subiculum. Prog Neurobiol 89:334–342. [DOI] [PubMed] [Google Scholar]
 - 14. Bender AT, Beavo JA (2006) Cyclic nucleotide phosphodiesterases: molecular regulation to clinical use. Pharmacol Rev 58:488–520. [DOI] [PubMed] [Google Scholar]
 - 15. Benito E, Barco A (2010) CREB's control of intrinsic and synaptic plasticity: implications for CREB‐dependent memory models. Trends Neurosci 33:230–240. [DOI] [PubMed] [Google Scholar]
 - 16. Berrios GE, Wagle AC, Markova IS, Wagle SA, Rosser A, Hodges JR (2002) Psychiatric symptoms in neurologically asymptomatic Huntington's disease gene carriers: a comparison with gene negative at risk subjects. Acta Psychiatr Scand 105:224–230. [DOI] [PubMed] [Google Scholar]
 - 17. Bezprozvanny I (2009) Calcium signaling and neurodegenerative diseases. Trends Mol Med 15:89–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 18. Bezprozvanny I (2011) Role of inositol 1,4,5‐trisphosphate receptors in pathogenesis of Huntington’ disease and spinocerebellar ataxias. Neurochem Res 36:1186–1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 19. Bibb JA, Yan Z, Svenningsson P, Snyder GL, Pieribone VA, Horiuchi A et al (2000) Severe deficiencies in dopamine signaling in presymptomatic Huntington's disease mice. Proc Natl Acad Sci USA 97:6809–6814. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 20. Bibel M, Hoppe E, Barde YA (1999) Biochemical and functional interactions between the neurotrophin receptors trk and p75NTR. Embo J 18:616–622. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 21. Bliss TV, Collingridge GL (1993) A synaptic model of memory: long‐term potentiation in the hippocampus. Nature 361:31–39. [DOI] [PubMed] [Google Scholar]
 - 22. Blokland A, Schreiber R, Prickaerts J (2006) Improving memory: a role for phosphodiesterases. Curr Pharm Des 12:2511–2523. [DOI] [PubMed] [Google Scholar]
 - 23. Boll TJ, Heaton R, Reitan RM (1974) Neuropsychological and emotional correlates of Huntington's chorea. J Nerv Ment Dis 158:61–69. [DOI] [PubMed] [Google Scholar]
 - 24. Boolell M, Allen MJ, Ballard SA, Gepi‐Attee S, Muirhead GJ, Naylor AM et al (1996) Sildenafil: an orally active type 5 cyclic GMP‐specific phosphodiesterase inhibitor for the treatment of penile erectile dysfunction. Int J Impot Res 8:47–52. [PubMed] [Google Scholar]
 - 25. Bourtchouladze R, Patterson SL, Kelly MP, Kreibich A, Kandel ER, Abel T (2006) Chronically increased Gsalpha signaling disrupts associative and spatial learning. Learn Mem 13:745–752. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 26. Bourtchuladze R, Frenguelli B, Blendy J, Cioffi D, Schutz G, Silva AJ (1994) Deficient long‐term memory in mice with a targeted mutation of the cAMP‐responsive element‐binding protein. Cell 79:59–68. [DOI] [PubMed] [Google Scholar]
 - 27. Bowles KR, Jones L (2014) Kinase signalling in Huntington's disease. J Huntingtons Dis 3:89–123. [DOI] [PubMed] [Google Scholar]
 - 28. Brito V, Giralt A, Enriquez‐Barreto L, Puigdellivol M, Suelves N, Zamora‐Moratalla A et al (2014) Neurotrophin receptor p75(NTR) mediates Huntington's disease‐associated synaptic and memory dysfunction. J Clin Invest 124:4411–4428. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 29. Brito V, Puigdellivol M, Giralt A, del TD, Alberch J, Gines S (2013) Imbalance of p75(NTR)/TrkB protein expression in Huntington’ disease: implication for neuroprotective therapies. Cell Death Dis 4:e595. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 30. Brooks S, Higgs G, Janghra N, Jones L, Dunnett SB (2012) Longitudinal analysis of the behavioural phenotype in YAC128 (C57BL/6J) Huntington’ disease transgenic mice. Brain Res Bull 88:113–120. [DOI] [PubMed] [Google Scholar]
 - 31. Brooks S, Higgs G, Jones L, Dunnett SB (2012) Longitudinal analysis of the behavioural phenotype in HdhQ92 Huntington's disease knock‐in mice. Brain Res Bull 88:148–155. [DOI] [PubMed] [Google Scholar]
 - 32. Brooks S, Higgs G, Jones L, Dunnett SB (2012) Longitudinal analysis of the behavioural phenotype in Hdh(CAG)150 Huntington’ disease knock‐in mice. Brain Res Bull 88:182–188. [DOI] [PubMed] [Google Scholar]
 - 33. Brooks SP, Betteridge H, Trueman RC, Jones L, Dunnett SB (2006) Selective extra‐dimensional set shifting deficit in a knock‐in mouse model of Huntington’ disease. Brain Res Bull 69:452–457. [DOI] [PubMed] [Google Scholar]
 - 34. Brooks SP, Dunnett SB (2013) Cognitive deficits in animal models of basal ganglia disorders. Brain Res Bull 92:29–40. [DOI] [PubMed] [Google Scholar]
 - 35. Brooks SP, Janghra N, Higgs GV, Bayram‐Weston Z, Heuer A, Jones L, Dunnett SB (2012) Selective cognitive impairment in the YAC128 Huntington’ disease mouse. Brain Res Bull 88:121–129. [DOI] [PubMed] [Google Scholar]
 - 36. Brooks SP, Janghra N, Workman VL, Bayram‐Weston Z, Jones L, Dunnett SB (2012) Longitudinal analysis of the behavioural phenotype in R6/1 (C57BL/6J) Huntington's disease transgenic mice. Brain Res Bull 88:94–103. [DOI] [PubMed] [Google Scholar]
 - 37. Buren C, Parsons MP, Smith‐Dijak A, Raymond LA (2016) Impaired development of cortico‐striatal synaptic connectivity in a cell culture model of Huntington's disease. Neurobiol Dis 87:80–90. [DOI] [PubMed] [Google Scholar]
 - 38. Burgess N, Maguire EA, O'Keefe J (2002) The human hippocampus and spatial and episodic memory. Neuron 35:625–641. [DOI] [PubMed] [Google Scholar]
 - 39. Cahill ME, Xie Z, Day M, Photowala H, Barbolina MV, Miller CA et al (2009) Kalirin regulates cortical spine morphogenesis and disease‐related behavioral phenotypes. Proc Natl Acad Sci USA 106:13058–13063. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 40. Caramins M, Halliday G, McCusker E, Trent RJ (2003) Genetically confirmed clinical Huntington's disease with no observable cell loss. J Neurol Neurosurg Psychiatry 74:968–970. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 41. Carter RJ, Lione LA, Humby T, Mangiarini L, Mahal A, Bates GP et al (1999) Characterization of progressive motor deficits in mice transgenic for the human Huntington's disease mutation. J Neurosci 19:3248–3257. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 42. Ceni C, Kommaddi RP, Thomas R, Vereker E, Liu X, McPherson PS et al (2010) The p75NTR intracellular domain generated by neurotrophin‐induced receptor cleavage potentiates Trk signaling. J Cell Sci 123:2299–2307. [DOI] [PubMed] [Google Scholar]
 - 43. Cepeda C, Cummings DM, Andre VM, Holley SM, Levine MS (2010) Genetic mouse models of Huntington's disease: focus on electrophysiological mechanisms. ASN Neurol 2:e00033. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 44. Cepeda C, Cummings DM, Hickey MA, Kleiman‐Weiner M, Chen JY, Watson JB, Levine MS (2010) Rescuing the corticostriatal synaptic disconnection in the R6/2 mouse model of Huntington’ Disease: exercise, adenosine receptors and ampakines. PLoS Curr 2:RRN1182. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 45. Cepeda C, Hurst RS, Calvert CR, Hernandez‐Echeagaray E, Nguyen OK, Jocoy E et al (2003) Transient and progressive electrophysiological alterations in the corticostriatal pathway in a mouse model of Huntington's disease. J Neurosci 23:961–969. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 46. Cepeda C, Starling AJ, Wu N, Nguyen OK, Uzgil B, Soda T et al (2004) Increased GABAergic function in mouse models of Huntington's disease: reversal by BDNF. J Neurosci Res 78:855–867. [DOI] [PubMed] [Google Scholar]
 - 47. Cepeda C, Wu N, Andre VM, Cummings DM, Levine MS (2007) The corticostriatal pathway in Huntington's disease. Prog Neurobiol 81:253–271. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 48. Chan HM, La Thangue NB (2001) p300/CBP proteins: HATs for transcriptional bridges and scaffolds. J Cell Sci 114:2363–2373. [DOI] [PubMed] [Google Scholar]
 - 49. Chang R, Liu X, Li S, Li XJ (2015) Transgenic animal models for study of the pathogenesis of Huntington's disease and therapy. Drug Des Dev Ther 9:2179–2188. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 50. Chaturvedi RK, Beal MF (2013) Mitochondria targeted therapeutic approaches in Parkinson's and Huntington's diseases. Mol Cell Neurosci 55:101–114. [DOI] [PubMed] [Google Scholar]
 - 51. Chen G, Zou X, Watanabe H, van Deursen JM, Shen J (2010) CREB binding protein is required for both short‐term and long‐term memory formation. J Neurosci 30:13066–13077. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 52. Chiba K, Adachi K (2012) Discovery of fingolimod, the sphingosine 1‐phosphate receptor modulator and its application for the therapy of multiple sclerosis. Future Med Chem 4:771–781. [DOI] [PubMed] [Google Scholar]
 - 53. Chrivia JC, Kwok RP, Lamb N, Hagiwara M, Montminy MR, Goodman RH (1993) Phosphorylated CREB binds specifically to the nuclear protein CBP. Nature 365:855–859. [DOI] [PubMed] [Google Scholar]
 - 54. Clarke JR, Cammarota M, Gruart A, Izquierdo I, Delgado‐Garcia JM (2010) Plastic modifications induced by object recognition memory processing. Proc Natl Acad Sci USA 107:2652–2657. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 55. Colomer V, Engelender S, Sharp AH, Duan K, Cooper JK, Lanahan A et al (1997) Huntingtin‐associated protein 1 (HAP1) binds to a Trio‐like polypeptide, with a rac1 guanine nucleotide exchange factor domain. Hum Mol Genet 6:1519–1525. [DOI] [PubMed] [Google Scholar]
 - 56. Coulson EJ, May LM, Osborne SL, Reid K, Underwood CK, Meunier FA et al (2008) p75 neurotrophin receptor mediates neuronal cell death by activating GIRK channels through phosphatidylinositol 4,5‐bisphosphate. J Neurosci 28:315–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 57. Cummings DM, Andre VM, Uzgil BO, Gee SM, Fisher YE, Cepeda C, Levine MS (2009) Alterations in cortical excitation and inhibition in genetic mouse models of Huntington's disease. J Neurosci 29:10371–10386. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 58. Cummings DM, Cepeda C, Levine MS (2010) Alterations in striatal synaptic transmission are consistent across genetic mouse models of Huntington's disease. ASN Neurol 2:e00036. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 59. Cummings DM, Milnerwood AJ, Dallerac GM, Vatsavayai SC, Hirst MC, Murphy KP (2007) Abnormal cortical synaptic plasticity in a mouse model of Huntington's disease. Brain Res Bull 72:103–107. [DOI] [PubMed] [Google Scholar]
 - 60. Cummings DM, Milnerwood AJ, Dallerac GM, Waights V, Brown JY, Vatsavayai SC et al (2006) Aberrant cortical synaptic plasticity and dopaminergic dysfunction in a mouse model of Huntington's disease. Hum Mol Genet 15:2856–2868. [DOI] [PubMed] [Google Scholar]
 - 61. Cybulska‐Klosowicz A, Mazarakis NK, van DA, Blakemore C, Hannan AJ, Kossut M (2004) Impaired learning‐dependent cortical plasticity in Huntington's disease transgenic mice. Neurobiol Dis 17:427–434. [DOI] [PubMed] [Google Scholar]
 - 62. Davies SW, Turmaine M, Cozens BA, DiFiglia M, Sharp AH, Ross CA et al (1997) Formation of neuronal intranuclear inclusions underlies the neurological dysfunction in mice transgenic for the HD mutation. Cell 90:537–548. [DOI] [PubMed] [Google Scholar]
 - 63. Deckel AW, Elder R, Fuhrer G (2002) Biphasic developmental changes in Ca2+/calmodulin‐dependent proteins in R6/2 Huntington's disease mice. Neuroreport 13:707–711. [DOI] [PubMed] [Google Scholar]
 - 64. Deckel AW, Gordinier A, Nuttal D, Tang V, Kuwada C, Freitas R, Gary KA (2001) Reduced activity and protein expression of NOS in R6/2 HD transgenic mice: effects of L‐NAME on symptom progression. Brain Res 919:70–81. [DOI] [PubMed] [Google Scholar]
 - 65. Deckel AW, Tang V, Nuttal D, Gary K, Elder R (2002) Altered neuronal nitric oxide synthase expression contributes to disease progression in Huntington's disease transgenic mice. Brain Res 939:76–86., [DOI] [PubMed] [Google Scholar]
 - 66. Deng YP, Wong T, Wan JY, Reiner A (2014) Differential loss of thalamostriatal and corticostriatal input to striatal projection neuron types prior to overt motor symptoms in the Q140 knock‐in mouse model of Huntington's disease. Front Syst Neurosci 8:198. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 67. Desplats PA, Kass KE, Gilmartin T, Stanwood GD, Woodward EL, Head SR et al (2006) Selective deficits in the expression of striatal‐enriched mRNAs in Huntington's disease. J Neurochem 96:743–757. [DOI] [PubMed] [Google Scholar]
 - 68. Di PA, Amico E, Favellato M, Castrataro R, Fucile S, Squitieri F, Maglione V (2014) FTY720 (fingolimod) is a neuroprotective and disease‐modifying agent in cellular and mouse models of Huntington disease. Hum Mol Genet 23:2251–2265. [DOI] [PubMed] [Google Scholar]
 - 69. Diaz‐Hernandez M, Torres‐Peraza J, Salvatori‐Abarca A, Moran MA, Gomez‐Ramos P, Alberch J, Lucas JJ (2005) Full motor recovery despite striatal neuron loss and formation of irreversible amyloid‐like inclusions in a conditional mouse model of Huntington's disease. J Neurosci 25:9773–9781. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 70. DiFiglia M, Sapp E, Chase KO, Davies SW, Bates GP, Vonsattel JP, Aronin N (1997) Aggregation of huntingtin in neuronal intranuclear inclusions and dystrophic neurites in brain. Science 277:1990–1993. [DOI] [PubMed] [Google Scholar]
 - 71. Dubnau J, Tully T (1998) Gene discovery in Drosophila: new insights for learning and memory. Annu Rev Neurosci 21:407–444. [DOI] [PubMed] [Google Scholar]
 - 72. Dumas EM, van den Bogaard SJ, Middelkoop HA, Roos RA (2013) A review of cognition in Huntington's disease. Front Biosci (Schol Ed) 5:1–18. [DOI] [PubMed] [Google Scholar]
 - 73. Ehrnhoefer DE, Sutton L, Hayden MR (2011) Small changes, big impact: posttranslational modifications and function of huntingtin in Huntington disease. Neuroscientist 17:475–492. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 74. Eichenbaum H (2001) The hippocampus and declarative memory: cognitive mechanisms and neural codes. Behav Brain Res 127:199–207. [DOI] [PubMed] [Google Scholar]
 - 75. Eichenbaum H (2004) Hippocampus: cognitive processes and neural representations that underlie declarative memory. Neuron 44:109–120. [DOI] [PubMed] [Google Scholar]
 - 76. Estrada‐Sanchez AM, Burroughs CL, Cavaliere S, Barton SJ, Chen S, Yang XW, Rebec GV (2015) Cortical efferents lacking mutant huntingtin improve striatal neuronal activity and behavior in a conditional mouse model of Huntington's disease. J Neurosci 35:4440–4451. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 77. Estrada‐Sanchez AM, Rebec GV (2013) Role of cerebral cortex in the neuropathology of Huntington's disease. Front Neural Circuits 7:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 78. Fan J, Gladding CM, Wang L, Zhang LY, Kaufman AM, Milnerwood AJ, Raymond LA (2012) P38 MAPK is involved in enhanced NMDA receptor‐dependent excitotoxicity in YAC transgenic mouse model of Huntington disease. Neurobiol Dis 45:999–1009. [DOI] [PubMed] [Google Scholar]
 - 79. Fan MM, Raymond LA (2007) N‐methyl‐d‐aspartate (NMDA) receptor function and excitotoxicity in Huntington's disease. Prog Neurobiol 81:272–293. [DOI] [PubMed] [Google Scholar]
 - 80. Fan MM, Zhang H, Hayden MR, Pelech SL, Raymond LA (2008) Protective up‐regulation of CK2 by mutant huntingtin in cells co‐expressing NMDA receptors. J Neurochem 104:790–805. [DOI] [PubMed] [Google Scholar]
 - 81. Ferrante RJ, Kowall NW, Richardson EP Jr. (1991) Proliferative and degenerative changes in striatal spiny neurons in Huntington's disease: a combined study using the section‐Golgi method and calbindin D28k immunocytochemistry. J Neurosci 11:3877–3887. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 82. Ferrante RJ, Kubilus JK, Lee J, Ryu H, Beesen A, Zucker B et al (2003) Histone deacetylase inhibition by sodium butyrate chemotherapy ameliorates the neurodegenerative phenotype in Huntington's disease mice. J Neurosci 23:9418–9427. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 83. Ferrer I, Goutan E, Marin C, Rey MJ, Ribalta T (2000) Brain‐derived neurotrophic factor in Huntington disease. Brain Res 866:257–261. [DOI] [PubMed] [Google Scholar]
 - 84. Ferreri K, Gill G, Montminy M (1994) The cAMP‐regulated transcription factor CREB interacts with a component of the TFIID complex. Proc Natl Acad Sci USA 91:1210–1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 85. Fossale E, Wheeler VC, Vrbanac V, Lebel LA, Teed A, Mysore JS et al (2002) Identification of a presymptomatic molecular phenotype in Hdh CAG knock‐in mice. Hum Mol Genet 11:2233–2241. [DOI] [PubMed] [Google Scholar]
 - 86. Friedman WJ (2000) Neurotrophins induce death of hippocampal neurons via the p75 receptor. J Neurosci 20:6340–6346. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 87. Gafni J, Hermel E, Young JE, Wellington CL, Hayden MR, Ellerby LM (2004) Inhibition of calpain cleavage of huntingtin reduces toxicity: accumulation of calpain/caspase fragments in the nucleus. J Biol Chem 279:20211–20220. [DOI] [PubMed] [Google Scholar]
 - 88. Gardian G, Browne SE, Choi DK, Klivenyi P, Gregorio J, Kubilus JK et al (2005) Neuroprotective effects of phenylbutyrate in the N171‐82Q transgenic mouse model of Huntington's disease. J Biol Chem 280:556–563. [DOI] [PubMed] [Google Scholar]
 - 89. Gharami K, Xie Y, An JJ, Tonegawa S, Xu B (2008) Brain‐derived neurotrophic factor over‐expression in the forebrain ameliorates Huntington's disease phenotypes in mice. J Neurochem 105:369–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 90. Ghilan M, Bostrom CA, Hryciw BN, Simpson JM, Christie BR, Gil‐Mohapel J (2014) YAC128 Huntington's disease transgenic mice show enhanced short‐term hippocampal synaptic plasticity early in the course of the disease. Brain Res 1581:117–128. [DOI] [PubMed] [Google Scholar]
 - 91. Giampa C, Laurenti D, Anzilotti S, Bernardi G, Menniti FS, Fusco FR (2010) Inhibition of the striatal specific phosphodiesterase PDE10A ameliorates striatal and cortical pathology in R6/2 mouse model of Huntington's disease. PLoS One 5:e13417. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 92. Giampa C, Patassini S, Borreca A, Laurenti D, Marullo F, Bernardi G et al (2009) Phosphodiesterase 10 inhibition reduces striatal excitotoxicity in the quinolinic acid model of Huntington's disease. Neurobiol Dis 34:450–456. [DOI] [PubMed] [Google Scholar]
 - 93. Gines S, Bosch M, Marco S, Gavalda N, Diaz‐Hernandez M, Lucas JJ et al (2006) Reduced expression of the TrkB receptor in Huntington’ disease mouse models and in human brain. Eur J Neurosci 23:649–658. [DOI] [PubMed] [Google Scholar]
 - 94. Gines S, Ivanova E, Seong IS, Saura CA, MacDonald ME (2003) Enhanced Akt signaling is an early pro‐survival response that reflects N‐methyl‐d‐aspartate receptor activation in Huntington's disease knock‐in striatal cells. J Biol Chem 278:50514–50522. [DOI] [PubMed] [Google Scholar]
 - 95. Gines S, Paoletti P, Alberch J (2010) Impaired TrkB‐mediated ERK1/2 activation in huntington disease knock‐in striatal cells involves reduced p52/p46 Shc expression. J Biol Chem 285:21537–21548. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 96. Gines S, Seong IS, Fossale E, Ivanova E, Trettel F, Gusella JF et al (2003) Specific progressive cAMP reduction implicates energy deficit in presymptomatic Huntington’ disease knock‐in mice. Hum Mol Genet 12:497–508. [DOI] [PubMed] [Google Scholar]
 - 97. Giralt A, Carreton O, Lao‐Peregrin C, Martin ED, Alberch J (2011) Conditional BDNF release under pathological conditions improves Huntington's disease pathology by delaying neuronal dysfunction. Mol Neurodegener 6:71. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 98. Giralt A, Puigdellivol M, Carreton O, Paoletti P, Valero J, Parra‐Damas A et al (2012) Long‐term memory deficits in Huntington's disease are associated with reduced CBP histone acetylase activity. Hum Mol Genet 21:1203–1216. [DOI] [PubMed] [Google Scholar]
 - 99. Giralt A, Rodrigo T, Martin ED, Gonzalez JR, Mila M, Cena V et al (2009) Brain‐derived neurotrophic factor modulates the severity of cognitive alterations induced by mutant huntingtin: involvement of phospholipaseCgamma activity and glutamate receptor expression. Neuroscience 158:1234–1250. [DOI] [PubMed] [Google Scholar]
 - 100. Giralt A, Saavedra A, Alberch J, Perez‐Navarro E (2012) Cognitive dysfunction in Huntington's disease: humans, mouse models and molecular mechanisms. J Huntingtons Dis 1:155–173. [DOI] [PubMed] [Google Scholar]
 - 101. Giralt A, Saavedra A, Carreton O, Arumi H, Tyebji S, Alberch J, Perez‐Navarro E (2013) PDE10 inhibition increases GluA1 and CREB phosphorylation and improves spatial and recognition memories in a Huntington's disease mouse model. Hippocampus 23:684–695. [DOI] [PubMed] [Google Scholar]
 - 102. Giralt A, Saavedra A, Carreton O, Xifro X, Alberch J, Perez‐Navarro E (2011) Increased PKA signaling disrupts recognition memory and spatial memory: role in Huntington's disease. Hum Mol Genet 20:4232–4247. [DOI] [PubMed] [Google Scholar]
 - 103. Gladding CM, Fitzjohn SM, Molnar E (2009) Metabotropic glutamate receptor‐mediated long‐term depression: molecular mechanisms. Pharmacol Rev 61:395–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 104. Gonzales ED, Tanenhaus AK, Zhang J, Chaffee RP, Yin JC (2016) Early‐onset sleep defects in Drosophila models of Huntington's disease reflect alterations of PKA/CREB signaling. Hum Mol Genet 25:837–852. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 105. Gorski JA, Zeiler SR, Tamowski S, Jones KR (2003) Brain‐derived neurotrophic factor is required for the maintenance of cortical dendrites. J Neurosci 23:6856–6865. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 106. Graveland GA, Williams RS, DiFiglia M (1985) Evidence for degenerative and regenerative changes in neostriatal spiny neurons in Huntington's disease. Science 227:770–773. [DOI] [PubMed] [Google Scholar]
 - 107. Gray M, Shirasaki DI, Cepeda C, Andre VM, Wilburn B, Lu XH et al (2008) Full‐length human mutant huntingtin with a stable polyglutamine repeat can elicit progressive and selective neuropathogenesis in BACHD mice. J Neurosci 28:6182–6195. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 108. Greferath U, Bennie A, Kourakis A, Bartlett PF, Murphy M, Barrett GL (2000) Enlarged cholinergic forebrain neurons and improved spatial learning in p75 knockout mice. Eur J Neurosci 12:885–893. [DOI] [PubMed] [Google Scholar]
 - 109. Gruart A, Delgado‐Garcia JM (2007) Activity‐dependent changes of the hippocampal CA3‐CA1 synapse during the acquisition of associative learning in conscious mice. Genes Brain Behav 6:24–31. [DOI] [PubMed] [Google Scholar]
 - 110. Gu X, Andre VM, Cepeda C, Li SH, Li XJ, Levine MS, Yang XW (2007) Pathological cell–cell interactions are necessary for striatal pathogenesis in a conditional mouse model of Huntington's disease. Mol Neurodegener 2:8. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 111. Gu X, Li C, Wei W, Lo V, Gong S, Li SH et al (2005) Pathological cell‐cell interactions elicited by a neuropathogenic form of mutant Huntingtin contribute to cortical pathogenesis in HD mice. Neuron 46:433–444. [DOI] [PubMed] [Google Scholar]
 - 112. Hahn‐Barma V, Deweer B, Durr A, Dode C, Feingold J, Pillon B et al (1998) Are cognitive changes the first symptoms of Huntington's disease? A study of gene carriers. J Neurol Neurosurg Psychiatry 64:172–177. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 113. Harrington DL, Smith MM, Zhang Y, Carlozzi NE, Paulsen JS (2012) Cognitive domains that predict time to diagnosis in prodromal Huntington disease. J Neurol Neurosurg Psychiatry 83:612–619. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 114. Hebb AL, Robertson HA (2007) Role of phosphodiesterases in neurological and psychiatric disease. Curr Opin Pharmacol 7:86–92. [DOI] [PubMed] [Google Scholar]
 - 115. Heck N, Betuing S, Vanhoutte P, Caboche J (2012) A deconvolution method to improve automated 3D‐analysis of dendritic spines: application to a mouse model of Huntington's disease. Brain Struct Funct 217:421–434. [DOI] [PubMed] [Google Scholar]
 - 116. Heckman PR, van Duinen MA, Bollen EP, Nishi A, Wennogle LP, Blokland A, Prickaerts J. Phosphodiesterase inhibition and regulation of dopaminergic frontal and striatal functioning: clinical implications. Int J Neuropsychopharmacol (in press) 2016 Apr 30. pii: pyw030. doi: 10.1093/ijnp/pyw030. [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 117. Herndon ES, Hladik CL, Shang P, Burns DK, Raisanen J, White CL III (2009) Neuroanatomic profile of polyglutamine immunoreactivity in Huntington disease brains. J Neuropathol Exp Neurol 68:250–261. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 118. Hickey MA, Kosmalska A, Enayati J, Cohen R, Zeitlin S, Levine MS, Chesselet MF (2008) Extensive early motor and non‐motor behavioral deficits are followed by striatal neuronal loss in knock‐in Huntington’ disease mice. Neuroscience 157:280–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 119. Ho AK, Sahakian BJ, Brown RG, Barker RA, Hodges JR, Ane MN et al (2003) Profile of cognitive progression in early Huntington's disease. Neurology 61:1702–1706. [DOI] [PubMed] [Google Scholar]
 - 120. Hodges A, Strand AD, Aragaki AK, Kuhn A, Sengstag T, Hughes G et al (2006) Regional and cellular gene expression changes in human Huntington's disease brain. Hum Mol Genet 15:965–977. [DOI] [PubMed] [Google Scholar]
 - 121. Hodgson JG, Agopyan N, Gutekunst CA, Leavitt BR, LePiane F, Singaraja R et al (1999) A YAC mouse model for Huntington's disease with full‐length mutant huntingtin, cytoplasmic toxicity, and selective striatal neurodegeneration. Neuron 23:181–192. [DOI] [PubMed] [Google Scholar]
 - 122. Hong SL, Cossyleon D, Hussain WA, Walker LJ, Barton SJ, Rebec GV (2012) Dysfunctional behavioral modulation of corticostriatal communication in the R6/2 mouse model of Huntington's disease. PLoS. One 7:e47026. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 123. Hult LS, Nilsson N, Soylu R, Kirik D, Petersen A (2013) Hypothalamic expression of mutant huntingtin contributes to the development of depressive‐like behavior in the BAC transgenic mouse model of Huntington's disease. Hum Mol Genet 22:3485–3497. [DOI] [PubMed] [Google Scholar]
 - 124. Ille R, Schafer A, Scharmuller W, Enzinger C, Schoggl H, Kapfhammer HP, Schienle A (2011) Emotion recognition and experience in Huntington disease: a voxel‐based morphometry study. J Psychiatry Neurosci 36:383–390. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 125. Jarabek BR, Yasuda RP, Wolfe BB (2004) Regulation of proteins affecting NMDA receptor‐induced excitotoxicity in a Huntington's mouse model. Brain 127:505–516. [DOI] [PubMed] [Google Scholar]
 - 126. Jia H, Morris CD, Williams RM, Loring JF, Thomas EA (2015) HDAC inhibition imparts beneficial transgenerational effects in Huntington's disease mice via altered DNA and histone methylation. Proc Natl Acad Sci USA 112:E56–E64. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 127. Jia H, Wang Y, Morris CD, Jacques V, Gottesfeld JM, Rusche JR, Thomas EA (2016) The effects of pharmacological inhibition of histone deacetylase 3 (HDAC3) in Huntington's disease mice. PLoS One 11:e0152498. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 128. Jiang H, Poirier MA, Liang Y, Pei Z, Weiskittel CE, Smith WW et al (2006) Depletion of CBP is directly linked with cellular toxicity caused by mutant huntingtin. Neurobiol Dis 23:543–551. [DOI] [PubMed] [Google Scholar]
 - 129. Joshi PR, Wu NP, Andre VM, Cummings DM, Cepeda C, Joyce JA et al (2009) Age‐dependent alterations of corticostriatal activity in the YAC128 mouse model of Huntington disease. J Neurosci 29:2414–2427. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 130. Kalkhoven E (2004) CBP and p300: HATs for different occasions. Biochem Pharmacol 68:1145–1155. [DOI] [PubMed] [Google Scholar]
 - 131. Kandel ER (2001) The molecular biology of memory storage: a dialogue between genes and synapses. Science 294:1030–1038. [DOI] [PubMed] [Google Scholar]
 - 132. Kandel ER (2012) The molecular biology of memory: cAMP, PKA, CRE, CREB‐1, CREB‐2, and CPEB. Mol Brain 5:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 133. Kandel ER, Pittenger C (1999) The past, the future and the biology of memory storage. Philos Trans R Soc Lond B Biol Sci 354:2027–2052. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 134. Kassubek J, Juengling FD, Kioschies T, Henkel K, Karitzky J, Kramer B et al (2004) Topography of cerebral atrophy in early Huntington's disease: a voxel based morphometric MRI study. J Neurol Neurosurg Psychiatry 75:213–220. [PMC free article] [PubMed] [Google Scholar]
 - 135. Kehoe LA, Bellone C, De RM, Zandueta A, Dey PN, Perez‐Otano I, Muller D (2014) GluN3A promotes dendritic spine pruning and destabilization during postnatal development. J Neurosci 34:9213–9221. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 136. Kelly MP, Cheung YF, Favilla C, Siegel SJ, Kanes SJ, Houslay MD, Abel T (2008) Constitutive activation of the G‐protein subunit Galphas within forebrain neurons causes PKA‐dependent alterations in fear conditioning and cortical Arc mRNA expression. Learn Mem 15:75–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 137. Kennedy MB, Beale HC, Carlisle HJ, Washburn LR (2005) Integration of biochemical signalling in spines. Nat Rev Neurosci 6:423–434. [DOI] [PubMed] [Google Scholar]
 - 138. Kessels RP, de Haan EH, Kappelle LJ, Postma A (2001) Varieties of human spatial memory: a meta‐analysis on the effects of hippocampal lesions. Brain Res Brain Res Rev 35:295–303. [DOI] [PubMed] [Google Scholar]
 - 139. Killoran A, Biglan KM (2014) Current therapeutic options for Huntington's disease: good clinical practice versus evidence‐based approaches? Mov Disord 29:1404–1413. [DOI] [PubMed] [Google Scholar]
 - 140. Kim JS, Reading SA, Brashers‐Krug T, Calhoun VD, Ross CA, Pearlson GD (2004) Functional MRI study of a serial reaction time task in Huntington's disease. Psychiatry Res 131:23–30. [DOI] [PubMed] [Google Scholar]
 - 141. Klapstein GJ, Fisher RS, Zanjani H, Cepeda C, Jokel ES, Chesselet MF, Levine MS (2001) Electrophysiological and morphological changes in striatal spiny neurons in R6/2 Huntington's disease transgenic mice. J Neurophysiol 86:2667–2677. [DOI] [PubMed] [Google Scholar]
 - 142. Kleiman RJ, Kimmel LH, Bove SE, Lanz TA, Harms JF, Romegialli A et al (2011) Chronic suppression of phosphodiesterase 10A alters striatal expression of genes responsible for neurotransmitter synthesis, neurotransmission, and signaling pathways implicated in Huntington's disease. J Pharmacol Exp Ther 336:64–76. [DOI] [PubMed] [Google Scholar]
 - 143. Kleppisch T, Feil R (2009) cGMP signalling in the mammalian brain: role in synaptic plasticity and behaviour. Handb Exp Pharmacol 191:549–579. [DOI] [PubMed] [Google Scholar]
 - 144. Korzus E, Rosenfeld MG, Mayford M (2004) CBP histone acetyltransferase activity is a critical component of memory consolidation. Neuron 42:961–972. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 145. Kumar A, Kumar SS, Kumar V, Kumar D, Agarwal S, Rana MK (2015) Huntington's disease: an update of therapeutic strategies. Gene 556:91–97. [DOI] [PubMed] [Google Scholar]
 - 146. Kwok RP, Lundblad JR, Chrivia JC, Richards JP, Bachinger HP, Brennan RG et al (1994) Nuclear protein CBP is a coactivator for the transcription factor CREB. Nature 370:223–226. [DOI] [PubMed] [Google Scholar]
 - 147. Labbadia J, Morimoto RI (2013) Huntington's disease: underlying molecular mechanisms and emerging concepts. Trends Biochem Sci 38:378–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 148. Laforet GA, Sapp E, Chase K, McIntyre C, Boyce FM, Campbell M et al (2001) Changes in cortical and striatal neurons predict behavioral and electrophysiological abnormalities in a transgenic murine model of Huntington's disease. J Neurosci 21:9112–9123. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 149. Lawrence AD, Hodges JR, Rosser AE, Kershaw A, ffrench‐Constant C, Rubinsztein DC et al (1998) Evidence for specific cognitive deficits in preclinical Huntington's disease. Brain 121:1329–1341. [DOI] [PubMed] [Google Scholar]
 - 150. Lawrence AD, Watkins LH, Sahakian BJ, Hodges JR, Robbins TW (2000) Visual object and visuospatial cognition in Huntington's disease: implications for information processing in corticostriatal circuits. Brain 123:1349–1364. [DOI] [PubMed] [Google Scholar]
 - 151. Lawrence AD, Weeks RA, Brooks DJ, Andrews TC, Watkins LH, Harding AE et al (1998) The relationship between striatal dopamine receptor binding and cognitive performance in Huntington's disease. Brain 121:1343–1355. [DOI] [PubMed] [Google Scholar]
 - 152. Lee B, Butcher GQ, Hoyt KR, Impey S, Obrietan K (2005) Activity‐dependent neuroprotection and cAMP response element‐binding protein (CREB): kinase coupling, stimulus intensity, and temporal regulation of CREB phosphorylation at serine 133. J Neurosci 25:1137–1148. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 153. Lee CF, Chern Y (2014) Adenosine receptors and Huntington's disease. Int Rev Neurobiol 119:195–232. [DOI] [PubMed] [Google Scholar]
 - 154. Lee J, Kim CH, Simon DK, Aminova LR, Andreyev AY, Kushnareva YE et al (2005) Mitochondrial cyclic AMP response element‐binding protein (CREB) mediates mitochondrial gene expression and neuronal survival. J Biol Chem 280:40398–40401. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 155. Lee ST, Chu K, Im WS, Yoon HJ, Im JY, Park JE et al (2011) Altered microRNA regulation in Huntington's disease models. Exp Neurol 227:172–179. [DOI] [PubMed] [Google Scholar]
 - 156. Lemiere J, Decruyenaere M, Evers‐Kiebooms G, Vandenbussche E, Dom R (2004) Cognitive changes in patients with Huntington's disease (HD) and asymptomatic carriers of the HD mutation—a longitudinal follow‐up study. J Neurol 251:935–942. [DOI] [PubMed] [Google Scholar]
 - 157. Lerner RP, Trejo Martinez LC, Zhu C, Chesselet MF, Hickey MA (2012) Striatal atrophy and dendritic alterations in a knock‐in mouse model of Huntington's disease. Brain Res Bull 87:571–578. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 158. Levine MS, Cepeda C, Hickey MA, Fleming SM, Chesselet MF (2004) Genetic mouse models of Huntington's and Parkinson's diseases: illuminating but imperfect. Trends Neurosci 27:691–697. [DOI] [PubMed] [Google Scholar]
 - 159. Levine MS, Klapstein GJ, Koppel A, Gruen E, Cepeda C, Vargas ME et al (1999) Enhanced sensitivity to N‐methyl‐d‐aspartate receptor activation in transgenic and knockin mouse models of Huntington's disease. J Neurosci Res 58:515–532. [PubMed] [Google Scholar]
 - 160. Li JY, Plomann M, Brundin P (2003) Huntington's disease: a synaptopathy? Trends Mol Med 9:414–420. [DOI] [PubMed] [Google Scholar]
 - 161. Li L, Fan M, Icton CD, Chen N, Leavitt BR, Hayden MR et al (2003) Role of NR2B‐type NMDA receptors in selective neurodegeneration in Huntington disease. Neurobiol Aging 24:1113–1121. [DOI] [PubMed] [Google Scholar]
 - 162. Li L, Murphy TH, Hayden MR, Raymond LA (2004) Enhanced striatal NR2B‐containing N‐methyl‐d‐aspartate receptor‐mediated synaptic currents in a mouse model of Huntington disease. J Neurophysiol 92:2738–2746. [DOI] [PubMed] [Google Scholar]
 - 163. Li SH, Cheng AL, Zhou H, Lam S, Rao M, Li H, Li XJ (2002) Interaction of Huntington disease protein with transcriptional activator Sp1. Mol Cell Biol 22:1277–1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 164. Li W, Silva HB, Real J, Wang YM, Rial D, Li P et al (2015) Inactivation of adenosine A2A receptors reverses working memory deficits at early stages of Huntington's disease models. Neurobiol Dis 79:70–80. [DOI] [PubMed] [Google Scholar]
 - 165. Lin CH, Tallaksen‐Greene S, Chien WM, Cearley JA, Jackson WS, Crouse AB et al (2001) Neurological abnormalities in a knock‐in mouse model of Huntington's disease. Hum Mol Genet 10:137–144. [DOI] [PubMed] [Google Scholar]
 - 166. Lione LA, Carter RJ, Hunt MJ, Bates GP, Morton AJ, Dunnett SB (1999) Selective discrimination learning impairments in mice expressing the human Huntington's disease mutation. J Neurosci 19:10428–10437. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 167. Lonze BE, Ginty DD (2002) Function and regulation of CREB family transcription factors in the nervous system. Neuron 35:605–623. [DOI] [PubMed] [Google Scholar]
 - 168. Lonze BE, Riccio A, Cohen S, Ginty DD (2002) Apoptosis, axonal growth defects, and degeneration of peripheral neurons in mice lacking CREB. Neuron 34:371–385. [DOI] [PubMed] [Google Scholar]
 - 169. Lu B, Pang PT, Woo NH (2005) The yin and yang of neurotrophin action. Nat Rev Neurosci 6:603–614. [DOI] [PubMed] [Google Scholar]
 - 170. Luthi‐Carter R, Hanson SA, Strand AD, Bergstrom DA, Chun W, Peters NL et al (2002) Dysregulation of gene expression in the R6/2 model of polyglutamine disease: parallel changes in muscle and brain. Hum Mol Genet 11:1911–1926. [DOI] [PubMed] [Google Scholar]
 - 171. Luthi‐Carter R, Strand A, Peters NL, Solano SM, Hollingsworth ZR, Menon AS et al (2000) Decreased expression of striatal signaling genes in a mouse model of Huntington's disease. Hum Mol Genet 9:1259–1271. [DOI] [PubMed] [Google Scholar]
 - 172. Lynch G, Kramar EA, Rex CS, Jia Y, Chappas D, Gall CM, Simmons DA (2007) Brain‐derived neurotrophic factor restores synaptic plasticity in a knock‐in mouse model of Huntington's disease. J Neurosci 27:4424–4434. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 173. Lynch MA (2004) Long‐term potentiation and memory. Physiol Rev 84:87–136. [DOI] [PubMed] [Google Scholar]
 - 174. Ma XM, Huang J, Wang Y, Eipper BA, Mains RE (2003) Kalirin, a multifunctional Rho guanine nucleotide exchange factor, is necessary for maintenance of hippocampal pyramidal neuron dendrites and dendritic spines. J Neurosci 23:10593–10603. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 175. Ma XM, Kiraly DD, Gaier ED, Wang Y, Kim EJ, Levine ES et al (2008) Kalirin‐7 is required for synaptic structure and function. J Neurosci 28:12368–12382. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 176. MacPhee IJ, Barker PA (1997) Brain‐derived neurotrophic factor binding to the p75 neurotrophin receptor reduces TrkA signaling while increasing serine phosphorylation in the TrkA intracellular domain. J Biol Chem 272:23547–23551. [DOI] [PubMed] [Google Scholar]
 - 177. Mangiarini L, Sathasivam K, Seller M, Cozens B, Harper A, Hetherington C et al (1996) Exon 1 of the HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice. Cell 87:493–506. [DOI] [PubMed] [Google Scholar]
 - 178. Mansuy IM, Shenolikar S (2006) Protein serine/threonine phosphatases in neuronal plasticity and disorders of learning and memory. Trends Neurosci 29:679–686. [DOI] [PubMed] [Google Scholar]
 - 179. Marco S, Giralt A, Petrovic MM, Pouladi MA, Martinez‐Turrillas R, Martinez‐Hernandez J et al (2013) Suppressing aberrant GluN3A expression rescues synaptic and behavioral impairments in Huntington's disease models. Nat Med 19:1030–1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 180. Marder K, Zhao H, Myers RH, Cudkowicz M, Kayson E, Kieburtz K et al (2000) Rate of functional decline in Huntington’ disease. Huntington Study Group. Neurology 54:452–458. [DOI] [PubMed] [Google Scholar]
 - 181. Marti E, Pantano L, Banez‐Coronel M, Llorens F, Minones‐Moyano E, Porta S et al (2010) A myriad of miRNA variants in control and Huntington's disease brain regions detected by massively parallel sequencing. Nucleic Acids Res 38:7219–7235. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 182. Mayford M (2007) Protein kinase signaling in synaptic plasticity and memory. Curr Opin Neurobiol 17:313–317. [DOI] [PubMed] [Google Scholar]
 - 183. Mazarakis NK, Cybulska‐Klosowicz A, Grote H, Pang T, van DA, Kossut M et al (2005) Deficits in experience‐dependent cortical plasticity and sensory‐discrimination learning in presymptomatic Huntington's disease mice. J Neurosci 25:3059–3066. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 184. McBride JL, Ramaswamy S, Gasmi M, Bartus RT, Herzog CD, Brandon EP et al (2006) Viral delivery of glial cell line‐derived neurotrophic factor improves behavior and protects striatal neurons in a mouse model of Huntington's disease. Proc Natl Acad Sci USA 103:9345–9350. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 185. McCampbell A, Taye AA, Whitty L, Penney E, Steffan JS, Fischbeck KH (2001) Histone deacetylase inhibitors reduce polyglutamine toxicity. Proc Natl Acad Sci USA 98:15179–15184. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 186. Meade CA, Deng YP, Fusco FR, Del MN, Hersch S, Goldowitz D, Reiner A (2002) Cellular localization and development of neuronal intranuclear inclusions in striatal and cortical neurons in R6/2 transgenic mice. J Comp Neurol 449:241–269. [DOI] [PubMed] [Google Scholar]
 - 187. Menalled L, Brunner D (2014) Animal models of Huntington's disease for translation to the clinic: best practices. Mov Disord 29:1375–1390. [DOI] [PubMed] [Google Scholar]
 - 188. Menalled LB, Sison JD, Dragatsis I, Zeitlin S, Chesselet MF (2003) Time course of early motor and neuropathological anomalies in a knock‐in mouse model of Huntington's disease with 140 CAG repeats. J Comp Neurol 465:11–26. [DOI] [PubMed] [Google Scholar]
 - 189. Metzler M, Gan L, Mazarei G, Graham RK, Liu L, Bissada N et al (2010) Phosphorylation of huntingtin at Ser421 in YAC128 neurons is associated with protection of YAC128 neurons from NMDA‐mediated excitotoxicity and is modulated by PP1 and PP2A. J Neurosci 30:14318–14329. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 190. Miguez A, Garcia‐Diaz BG, Brito V, Straccia M, Giralt A, Gines S et al (2015) Fingolimod (FTY720) enhances hippocampal synaptic plasticity and memory in Huntington's disease by preventing p75NTR up‐regulation and astrocyte‐mediated inflammation. Hum Mol Genet 24:4958–4970. [DOI] [PubMed] [Google Scholar]
 - 191. Miller BR, Bezprozvanny I (2010) Corticostriatal circuit dysfunction in Huntington’ disease: intersection of glutamate, dopamine and calcium. Future Neurol 5:735–756. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 192. Milnerwood AJ, Cummings DM, Dallerac GM, Brown JY, Vatsavayai SC, Hirst MC et al (2006) Early development of aberrant synaptic plasticity in a mouse model of Huntington's disease. Hum Mol Genet 15:1690–1703. [DOI] [PubMed] [Google Scholar]
 - 193. Milnerwood AJ, Gladding CM, Pouladi MA, Kaufman AM, Hines RM, Boyd JD et al (2010) Early increase in extrasynaptic NMDA receptor signaling and expression contributes to phenotype onset in Huntington's disease mice. Neuron 65:178–190. [DOI] [PubMed] [Google Scholar]
 - 194. Milnerwood AJ, Raymond LA (2007) Corticostriatal synaptic function in mouse models of Huntington's disease: early effects of huntingtin repeat length and protein load. J Physiol 585:817–831. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 195. Mitsuishi M, Miyashita K, Itoh H (2008) cGMP rescues mitochondrial dysfunction induced by glucose and insulin in myocytes. Biochem Biophys Res Commun 367:840–845. [DOI] [PubMed] [Google Scholar]
 - 196. Montaldi D, Mayes AR (2010) The role of recollection and familiarity in the functional differentiation of the medial temporal lobes. Hippocampus 20:1291–1314. [DOI] [PubMed] [Google Scholar]
 - 197. Montoya A, Pelletier M, Menear M, Duplessis E, Richer F, Lepage M (2006) Episodic memory impairment in Huntington's disease: a meta‐analysis. Neuropsychologia 44:1984–1994. [DOI] [PubMed] [Google Scholar]
 - 198. Morton AJ (2013) Circadian and sleep disorder in Huntington's disease. Exp Neurol 243:34–44. [DOI] [PubMed] [Google Scholar]
 - 199. Morton AJ, Lagan MA, Skepper JN, Dunnett SB (2000) Progressive formation of inclusions in the striatum and hippocampus of mice transgenic for the human Huntington's disease mutation. J Neurocytol 29:679–702. [DOI] [PubMed] [Google Scholar]
 - 200. Murmu RP, Li W, Holtmaat A, Li JY (2013) Dendritic spine instability leads to progressive neocortical spine loss in a mouse model of Huntington's disease. J Neurosci 33:12997–13009. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 201. Murphy KP, Carter RJ, Lione LA, Mangiarini L, Mahal A, Bates GP 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 202. Nguyen PV, Woo NH (2003) Regulation of hippocampal synaptic plasticity by cyclic AMP‐dependent protein kinases. Prog Neurobiol 71:401–437. [DOI] [PubMed] [Google Scholar]
 - 203. Niccolini F, Haider S, Reis MT, Muhlert N, Tziortzi AC, Searle GE et al (2015) Altered PDE10A expression detectable early before symptomatic onset in Huntington's disease. Brain 138:3016–3029. [DOI] [PubMed] [Google Scholar]
 - 204. Nithianantharajah J, Hannan AJ (2013) Dysregulation of synaptic proteins, dendritic spine abnormalities and pathological plasticity of synapses as experience‐dependent mediators of cognitive and psychiatric symptoms in Huntington's disease. Neuroscience 251:66–74. [DOI] [PubMed] [Google Scholar]
 - 205. Norris PJ, Waldvogel HJ, Faull RL, Love DR, Emson PC (1996) Decreased neuronal nitric oxide synthase messenger RNA and somatostatin messenger RNA in the striatum of Huntington's disease. Neuroscience 72:1037–1047. [DOI] [PubMed] [Google Scholar]
 - 206. Nucifora FC Jr, Sasaki M, Peters MF, Huang H, Cooper JK, Yamada M et al (2001) Interference by huntingtin and atrophin‐1 with cbp‐mediated transcription leading to cellular toxicity. Science 291:2423–2428. [DOI] [PubMed] [Google Scholar]
 - 207. Numakawa T, Yokomaku D, Richards M, Hori H, Adachi N, Kunugi H (2010) Functional interactions between steroid hormones and neurotrophin BDNF. World J Biol Chem 1:133–143. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 208. Ogryzko VV, Schiltz RL, Russanova V, Howard BH, Nakatani Y (1996) The transcriptional coactivators p300 and CBP are histone acetyltransferases. Cell 87:953–959. [DOI] [PubMed] [Google Scholar]
 - 209. Oike Y, Hata A, Mamiya T, Kaname T, Noda Y, Suzuki M et al (1999) Truncated CBP protein leads to classical Rubinstein‐Taybi syndrome phenotypes in mice: implications for a dominant‐negative mechanism. Hum Mol Genet 8:387–396. [DOI] [PubMed] [Google Scholar]
 - 210. Orth M, Schippling S, Schneider SA, Bhatia KP, Talelli P, Tabrizi SJ, Rothwell JC (2010) Abnormal motor cortex plasticity in premanifest and very early manifest Huntington disease. J Neurol Neurosurg Psychiatry 81:267–270. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 211. Padovan‐Neto FE, Sammut S, Chakroborty S, Dec AM, Threlfell S, Campbell PW et al, (2015) Facilitation of corticostriatal transmission following pharmacological inhibition of striatal phosphodiesterase 10A: role of nitric oxide‐soluble guanylyl cyclase‐cGMP signaling pathways. J Neurosci 35:5781–5791. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 212. Paoletti P, Vila I, Rife M, Lizcano JM, Alberch J, Gines S (2008) Dopaminergic and glutamatergic signaling crosstalk in Huntington's disease neurodegeneration: the role of p25/cyclin‐dependent kinase 5. J Neurosci 28:10090–10101. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 213. Papoutsi M, Labuschagne I, Tabrizi SJ, Stout JC (2014) The cognitive burden in Huntington's disease: pathology, phenotype, and mechanisms of compensation. Mov Disord 29:673–683. [DOI] [PubMed] [Google Scholar]
 - 214. Papp KV, Kaplan RF, Snyder PJ (2011) Biological markers of cognition in prodromal Huntington’ disease: a review. Brain Cogn 77:280–291. [DOI] [PubMed] [Google Scholar]
 - 215. Parker D, Ferreri K, Nakajima T, LaMorte VJ, Evans R, Koerber SC et al (1996) Phosphorylation of CREB at Ser‐133 induces complex formation with CREB‐binding protein via a direct mechanism. Mol Cell Biol 16:694–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 216. Parker JA, Metzler M, Georgiou J, Mage M, Roder JC, Rose AM et al (2007) Huntingtin‐interacting protein 1 influences worm and mouse presynaptic function and protects Caenorhabditis elegans neurons against mutant polyglutamine toxicity. J Neurosci 27:11056–11064. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 217. Paulsen JS (2011) Cognitive impairment in Huntington disease: diagnosis and treatment. Curr Neurol Neurosci Rep 11:474–483. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 218. Paulsen JS, Butters N, Sadek JR, Johnson SA, Salmon DP, Swerdlow NR, Swenson MR (1995) Distinct cognitive profiles of cortical and subcortical dementia in advanced illness. Neurology 45:951–956. [DOI] [PubMed] [Google Scholar]
 - 219. Paulsen JS, Zhao H, Stout JC, Brinkman RR, Guttman M, Ross CA et al (2001) Clinical markers of early disease in persons near onset of Huntington's disease. Neurology 57:658–662. [DOI] [PubMed] [Google Scholar]
 - 220. Peixoto CA, Nunes AK, Garcia‐Osta A (2015) Phosphodiesterase‐5 inhibitors: action on the signaling pathways of neuroinflammation, neurodegeneration, and cognition. Mediators Inflamm 2015:940207. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 221. Penzes P, Johnson RC, Kambampati V, Mains RE, Eipper BA (2001) Distinct roles for the two Rho GDP/GTP exchange factor domains of kalirin in regulation of neurite growth and neuronal morphology. J Neurosci 21:8426–8434. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 222. Penzes P, Johnson RC, Sattler R, Zhang X, Huganir RL, Kambampati V et al (2001) The neuronal Rho‐GEF Kalirin‐7 interacts with PDZ domain‐containing proteins and regulates dendritic morphogenesis. Neuron 29:229–242. [DOI] [PubMed] [Google Scholar]
 - 223. Penzes P, Jones KA (2008) Dendritic spine dynamics–a key role for kalirin‐7. Trends Neurosci 31:419–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 224. Perez‐Otano I, Lujan R, Tavalin SJ, Plomann M, Modregger J, Liu XB et al (2006) Endocytosis and synaptic removal of NR3A‐containing NMDA receptors by PACSIN1/syndapin1. Nat Neurosci 9:611–621. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 225. Perez‐Severiano F, Escalante B, Vergara P, Rios C, Segovia J (2002) Age‐dependent changes in nitric oxide synthase activity and protein expression in striata of mice transgenic for the Huntington's disease mutation. Brain Res 951:36–42. [DOI] [PubMed] [Google Scholar]
 - 226. Pineda JR, Pardo R, Zala D, Yu H, Humbert S, Saudou F (2009) Genetic and pharmacological inhibition of calcineurin corrects the BDNF transport defect in Huntington's disease. Mol Brain 2:33. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 227. Plotkin JL, Day M, Peterson JD, Xie Z, Kress GJ, Rafalovich I et al (2014) Impaired TrkB receptor signaling underlies corticostriatal dysfunction in Huntington’ disease. Neuron 83:178–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 228. Plotkin JL, Surmeier DJ (2015) Corticostriatal synaptic adaptations in Huntington's disease. Curr Opin Neurobiol 33:53–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 229. Politis M, Pavese N, Tai YF, Tabrizi SJ, Barker RA, Piccini P (2008) Hypothalamic involvement in Huntington's disease: an in vivo PET study. Brain 131:2860–2869. [DOI] [PubMed] [Google Scholar]
 - 230. Poo MM (2001) Neurotrophins as synaptic modulators. Nat Rev Neurosci 2:24–32. [DOI] [PubMed] [Google Scholar]
 - 231. Popoli P (2008) Regulation of brain functions by A2A receptors: implication for therapeutics. Curr Pharm Des 14:1466–1467. [DOI] [PubMed] [Google Scholar]
 - 232. Popoli P, Blum D, Martire A, Ledent C, Ceruti S, Abbracchio MP (2007) Functions, dysfunctions and possible therapeutic relevance of adenosine A2A receptors in Huntington's disease. Prog Neurobiol 81:331–348. [DOI] [PubMed] [Google Scholar]
 - 233. Popugaeva E, Pchitskaya E, Speshilova A, Alexandrov S, Zhang H, Vlasova O, Bezprozvanny I (2015) STIM2 protects hippocampal mushroom spines from amyloid synaptotoxicity. Mol Neurodegener 10:37. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 234. Poudel GR, Egan GF, Churchyard A, Chua P, Stout JC, Georgiou‐Karistianis N (2014) Abnormal synchrony of resting state networks in premanifest and symptomatic Huntington disease: the IMAGE‐HD study. J Psychiatry Neurosci 39:87–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 235. Poudel GR, Stout JC, Dominguez DJ, Salmon L, Churchyard A, Chua P et al (2014) White matter connectivity reflects clinical and cognitive status in Huntington's disease. Neurobiol Dis 65:180–187. [DOI] [PubMed] [Google Scholar]
 - 236. Pouladi MA, Stanek LM, Xie Y, Franciosi S, Southwell AL, Deng Y et al (2012) Marked differences in neurochemistry and aggregates despite similar behavioural and neuropathological features of Huntington disease in the full‐length BACHD and YAC128 mice. Hum Mol Genet 21:2219–2232. [DOI] [PubMed] [Google Scholar]
 - 237. Pouladi MA, Xie Y, Skotte NH, Ehrnhoefer DE, Graham RK, Kim JE et al (2010) Full‐length huntingtin levels modulate body weight by influencing insulin‐like growth factor 1 expression. Hum Mol Genet 19:1528–1538. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 238. Puerta E, Hervias I, Barros‐Minones L, Jordan J, Ricobaraza A, Cuadrado‐Tejedor M et al (2010) Sildenafil protects against 3‐nitropropionic acid neurotoxicity through the modulation of calpain, CREB, and BDNF. Neurobiol Dis 38:237–245. [DOI] [PubMed] [Google Scholar]
 - 239. Puigdellivol M, Cherubini M, Brito V, Giralt A, Suelves N, Ballesteros J et al (2015) A role for Kalirin‐7 in corticostriatal synaptic dysfunction in Huntington's disease. Hum Mol Genet 24:7265–7285. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 240. Putney JW Jr (2003) Capacitative calcium entry in the nervous system. Cell Calcium 34:339–344. [DOI] [PubMed] [Google Scholar]
 - 241. Puzzo D, Sapienza S, Arancio O, Palmeri A (2008) Role of phosphodiesterase 5 in synaptic plasticity and memory. Neuropsychiatr Dis Treat 4:371–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 242. Radhakrishnan I, Perez‐Alvarado GC, Parker D, Dyson HJ, Montminy MR, Wright PE (1997) Solution structure of the KIX domain of CBP bound to the transactivation domain of CREB: a model for activator:coactivator interactions. Cell 91:741–752. [DOI] [PubMed] [Google Scholar]
 - 243. Ramaswamy S, McBride JL, Kordower JH (2007) Animal models of Huntington's disease. ILAR J 48:356–373. [DOI] [PubMed] [Google Scholar]
 - 244. Ravnskjaer K, Kester H, Liu Y, Zhang X, Lee D, Yates JR III, Montminy M (2007) Cooperative interactions between CBP and TORC2 confer selectivity to CREB target gene expression. Embo J 26:2880–2889. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 245. Raymond LA, Andre VM, Cepeda C, Gladding CM, Milnerwood AJ, Levine MS (2011) Pathophysiology of Huntington's disease: time‐dependent alterations in synaptic and receptor function. Neuroscience 198:252–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 246. Reiner A, Albin RL, Anderson KD, D'Amato CJ, Penney JB, Young AB (1988) Differential loss of striatal projection neurons in Huntington disease. Proc Natl Acad Sci USA 85:5733–5737. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 247. Roberts AC, Diez‐Garcia J, Rodriguiz RM, Lopez IP, Lujan R, Martinez‐Turrillas R et al (2009) Downregulation of NR3A‐containing NMDARs is required for synapse maturation and memory consolidation. Neuron 63:342–356. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 248. Rocher AB, Gubellini P, Merienne N, Boussicault L, Petit F, Gipchtein P et al (2016) Synaptic scaling up in medium spiny neurons of aged BACHD mice: a slow‐progression model of Huntington's disease. Neurobiol Dis 86:131–139. [DOI] [PubMed] [Google Scholar]
 - 249. Rosas HD, Koroshetz WJ, Chen YI, Skeuse C, Vangel M, Cudkowicz ME et al (2003) Evidence for more widespread cerebral pathology in early HD: an MRI‐based morphometric analysis. Neurology 60:1615–1620. [DOI] [PubMed] [Google Scholar]
 - 250. Rosas HD, Lee SY, Bender AC, Zaleta AK, Vangel M, Yu P et al (2010) Altered white matter microstructure in the corpus callosum in Huntington's disease: implications for cortical “disconnection”. Neuroimage 49:2995–3004. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 251. Rosch H, Schweigreiter R, Bonhoeffer T, Barde YA, Korte M (2005) The neurotrophin receptor p75NTR modulates long‐term depression and regulates the expression of AMPA receptor subunits in the hippocampus. Proc Natl Acad Sci USA 102:7362–7367. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 252. Rosenberg NK, Sorensen SA, Christensen AL (1995) Neuropsychological characteristics of Huntington's disease carriers: a double blind study. J Med Genet 32:600–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 253. Rosenfeld MG, Glass CK (2001) Coregulator codes of transcriptional regulation by nuclear receptors. J Biol Chem 276:36865–36868. [DOI] [PubMed] [Google Scholar]
 - 254. Rossi S, Prosperetti C, Picconi B, De Chiara V, Mataluni G, Bernardi G et al (2006) Deficits of glutamate transmission in the striatum of toxic and genetic models of Huntington's disease. Neurosci Lett 410:6–10. [DOI] [PubMed] [Google Scholar]
 - 255. Rouaux C, Loeffler JP, Boutillier AL (2004) Targeting CREB‐binding protein (CBP) loss of function as a therapeutic strategy in neurological disorders. Biochem Pharmacol 68:1157–1164. [DOI] [PubMed] [Google Scholar]
 - 256. Roze E, Betuing S, Deyts C, Marcon E, Brami‐Cherrier K, Pages C et al (2008) Mitogen‐ and stress‐activated protein kinase‐1 deficiency is involved in expanded‐huntingtin‐induced transcriptional dysregulation and striatal death. FASEB J 22:1083–1093., [DOI] [PubMed] [Google Scholar]
 - 257. Rue L, Alcala‐Vida R, Lopez‐Soop G, Creus‐Muncunill J, Alberch J, Perez‐Navarro E (2014) Early down‐regulation of PKCdelta as a pro‐survival mechanism in Huntington's disease. Neuromolecular Med 16:25–37. [DOI] [PubMed] [Google Scholar]
 - 258. Runne H, Regulier E, Kuhn A, Zala D, Gokce O, Perrin V et al (2008) Dysregulation of gene expression in primary neuron models of Huntington's disease shows that polyglutamine‐related effects on the striatal transcriptome may not be dependent on brain circuitry. J Neurosci 28:9723–9731. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 259. Russell DS, Jennings DL, Barret O, Tamagnan GD, Carroll VM, Caille F et al (2016) Change in PDE10 across early Huntington disease assessed by [18F]MNI‐659 and PET imaging. Neurology 86:748–754. [DOI] [PubMed] [Google Scholar]
 - 260. Saavedra A, Garcia‐Martinez JM, Xifro X, Giralt A, Torres‐Peraza JF, Canals JM et al (2010) PH domain leucine‐rich repeat protein phosphatase 1 contributes to maintain the activation of the PI3K/Akt pro‐survival pathway in Huntington's disease striatum. Cell Death Differ 17:324–335. [DOI] [PubMed] [Google Scholar]
 - 261. Saavedra A, Giralt A, Arumi H, Alberch J, Perez‐Navarro E (2013) Regulation of hippocampal cGMP levels as a candidate to treat cognitive deficits in Huntington's disease. PLoS One 8:e73664. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 262. Saavedra A, Giralt A, Rue L, Xifro X, Xu J, Ortega Z et al (2011) Striatal‐enriched protein tyrosine phosphatase expression and activity in Huntington's disease: a STEP in the resistance to excitotoxicity. J Neurosci 31:8150–8162. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 263. Sadri‐Vakili G, Bouzou B, Benn CL, Kim MO, Chawla P, Overland RP et al, (2007) Histones associated with downregulated genes are hypo‐acetylated in Huntington's disease models. Hum Mol Genet 16:1293–1306. [DOI] [PubMed] [Google Scholar]
 - 264. Sapp E, Penney J, Young A, Aronin N, Vonsattel JP, DiFiglia M (1999) Axonal transport of N‐terminal huntingtin suggests early pathology of corticostriatal projections in Huntington disease. J Neuropathol Exp Neurol 58:165–173. [DOI] [PubMed] [Google Scholar]
 - 265. Sapp E, Schwarz C, Chase K, Bhide PG, Young AB, Penney J et al (1997) Huntingtin localization in brains of normal and Huntington's disease patients. Ann Neurol 42:604–612. [DOI] [PubMed] [Google Scholar]
 - 266. Saura CA, Choi SY, Beglopoulos V, Malkani S, Zhang D, Shankaranarayana Rao BS et al (2004) Loss of presenilin function causes impairments of memory and synaptic plasticity followed by age‐dependent neurodegeneration. Neuron 42:23–36. [DOI] [PubMed] [Google Scholar]
 - 267. Schaffar G, Breuer P, Boteva R, Behrends C, Tzvetkov N, Strippel N et al (2004) Cellular toxicity of polyglutamine expansion proteins: mechanism of transcription factor deactivation. Mol Cell 15:95–105. [DOI] [PubMed] [Google Scholar]
 - 268. Schilling G, Becher MW, Sharp AH, Jinnah HA, Duan K, Kotzuk JA et al (1999) Intranuclear inclusions and neuritic aggregates in transgenic mice expressing a mutant N‐terminal fragment of huntingtin. Hum Mol Genet 8:397–407. [DOI] [PubMed] [Google Scholar]
 - 269. Schippling S, Schneider SA, Bhatia KP, Munchau A, Rothwell JC, Tabrizi SJ, Orth M (2009) Abnormal motor cortex excitability in preclinical and very early Huntington's disease. Biol Psychiatry 65:959–965. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 270. Schneider SA, Wilkinson L, Bhatia KP, Henley SM, Rothwell JC, Tabrizi SJ, Jahanshahi M (2010) Abnormal explicit but normal implicit sequence learning in premanifest and early Huntington's disease. Mov Disord 25:1343–1349. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 271. Seo H, Kim W, Isacson O (2008) Compensatory changes in the ubiquitin‐proteasome system, brain‐derived neurotrophic factor and mitochondrial complex II/III in YAC72 and R6/2 transgenic mice partially model Huntington's disease patients. Hum Mol Genet 17:3144–3153. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 272. Sepers MD, Raymond LA (2014) Mechanisms of synaptic dysfunction and excitotoxicity in Huntington's disease. Drug Discov Today 19:990–996. [DOI] [PubMed] [Google Scholar]
 - 273. Seredenina T, Luthi‐Carter R (2012) What have we learned from gene expression profiles in Huntington's disease? Neurobiol Dis 45:83–98. [DOI] [PubMed] [Google Scholar]
 - 274. Shepherd GM, Erulkar SD (1997) Centenary of the synapse: from Sherrington to the molecular biology of the synapse and beyond. Trends Neurosci 20:385–392. [DOI] [PubMed] [Google Scholar]
 - 275. Shirasaki DI, Greiner ER, Al‐Ramahi I, Gray M, Boontheung P, Geschwind DH et al (2012) Network organization of the huntingtin proteomic interactome in mammalian brain. Neuron 75:41–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 276. Silva AJ, Kogan JH, Frankland PW, Kida S (1998) CREB and memory. Annu Rev Neurosci 21:127–148. [DOI] [PubMed] [Google Scholar]
 - 277. Simmons DA, Mehta RA, Lauterborn JC, Gall CM, Lynch G (2011) Brief ampakine treatments slow the progression of Huntington's disease phenotypes in R6/2 mice. Neurobiol Dis 41:436–444. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 278. Siuciak JA (2008) The role of phosphodiesterases in schizophrenia: therapeutic implications. CNS Drugs 22:983–993. [DOI] [PubMed] [Google Scholar]
 - 279. Skaper SD (2012) The neurotrophin family of neurotrophic factors: an overview. Methods Mol Biol 846:1–12. [DOI] [PubMed] [Google Scholar]
 - 280. Slow EJ, van RJ, Rogers D, Coleman SH, Graham RK, Deng Y et al (2003) Selective striatal neuronal loss in a YAC128 mouse model of Huntington disease. Hum Mol Genet 12:1555–1567. [DOI] [PubMed] [Google Scholar]
 - 281. Smith R, Brundin P, Li JY (2005) Synaptic dysfunction in Huntington's disease: a new perspective. Cell Mol Life Sci 62:1901–1912. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 282. Song W, Volosin M, Cragnolini AB, Hempstead BL, Friedman WJ (2010) ProNGF induces PTEN via p75NTR to suppress Trk‐mediated survival signaling in brain neurons. J Neurosci 30:15608–15615. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 283. Sotrel A, Williams RS, Kaufmann WE, Myers RH (1993) Evidence for neuronal degeneration and dendritic plasticity in cortical pyramidal neurons of Huntington's disease: a quantitative Golgi study. Neurology 43:2088–2096. [DOI] [PubMed] [Google Scholar]
 - 284. Spampanato J, Gu X, Yang XW, Mody I (2008) Progressive synaptic pathology of motor cortical neurons in a BAC transgenic mouse model of Huntington's disease. Neuroscience 157:606–620. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 285. Spargo E, Everall IP, Lantos PL (1993) Neuronal loss in the hippocampus in Huntington's disease: a comparison with HIV infection. J Neurol Neurosurg Psychiatry 56:487–491. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 286. Spires TL, Grote HE, Garry S, Cordery PM, van DA, Blakemore C, Hannan AJ (2004) Dendritic spine pathology and deficits in experience‐dependent dendritic plasticity in R6/1 Huntington's disease transgenic mice. Eur J Neurosci 19:2799–2807. [DOI] [PubMed] [Google Scholar]
 - 287. Stack EC, Kubilus JK, Smith K, Cormier K, Del Signore SJ, Guelin E et al (2005) Chronology of behavioral symptoms and neuropathological sequela in R6/2 Huntington's disease transgenic mice. J Comp Neurol 490:354–370. [DOI] [PubMed] [Google Scholar]
 - 288. Steffan JS, Bodai L, Pallos J, Poelman M, McCampbell A, Apostol BL et al (2001) Histone deacetylase inhibitors arrest polyglutamine‐dependent neurodegeneration in Drosophila. Nature 413:739–743. [DOI] [PubMed] [Google Scholar]
 - 289. Steffan JS, Kazantsev A, Spasic‐Boskovic O, Greenwald M, Zhu YZ, Gohler H et al, (2000) The Huntington's disease protein interacts with p53 and CREB‐binding protein and represses transcription. Proc Natl Acad Sci USA 97:6763–6768. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 290. Stout JC, Glikmann‐Johnston Y, Andrews SC (2016) Cognitive assessment strategies in Huntington's disease research. J Neurosci Methods 265:19–24. [DOI] [PubMed] [Google Scholar]
 - 291. Stout JC, Paulsen JS, Queller S, Solomon AC, Whitlock KB, Campbell JC et al (2011) Neurocognitive signs in prodromal Huntington disease. Neuropsychology 25:1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 292. Sugars KL, Brown R, Cook LJ, Swartz J, Rubinsztein DC (2004) Decreased cAMP response element‐mediated transcription: an early event in exon 1 and full‐length cell models of Huntington's disease that contributes to polyglutamine pathogenesis. J Biol Chem 279:4988–4999. [DOI] [PubMed] [Google Scholar]
 - 293. Sun S, Zhang H, Liu J, Popugaeva E, Xu NJ, Feske S et al (2014) Reduced synaptic STIM2 expression and impaired store‐operated calcium entry cause destabilization of mature spines in mutant presenilin mice. Neuron 82:79–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 294. Suopanki J, Gotz C, Lutsch G, Schiller J, Harjes P, Herrmann A, Wanker EE (2006) Interaction of huntingtin fragments with brain membranes—clues to early dysfunction in Huntington's disease. J Neurochem 96:870–884. [DOI] [PubMed] [Google Scholar]
 - 295. Tang TS, Slow E, Lupu V, Stavrovskaya IG, Sugimori M, Llinas R et al (2005) Disturbed Ca2+ signaling and apoptosis of medium spiny neurons in Huntington’ disease. Proc Natl Acad Sci USA 102:2602–2607. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 296. Tang TS, Tu H, Chan EY, Maximov A, Wang Z, Wellington CL et al (2003) Huntingtin and huntingtin‐associated protein 1 influence neuronal calcium signaling mediated by inositol‐(1,4,5) triphosphate receptor type 1. Neuron 39:227–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 297. Taylor JP, Taye AA, Campbell C, Kazemi‐Esfarjani P, Fischbeck KH, Min KT (2003) Aberrant histone acetylation, altered transcription, and retinal degeneration in a Drosophila model of polyglutamine disease are rescued by CREB‐binding protein. Genes Dev 17:1463–1468. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 298. Thakur T, Sharma S, Kumar K, Deshmukh R, Sharma PL (2013) Neuroprotective role of PDE4 and PDE5 inhibitors in 3‐nitropropionic acid induced behavioral and biochemical toxicities in rats. Eur J Pharmacol 714:515–521. [DOI] [PubMed] [Google Scholar]
 - 299. The Huntington's Disease Collaborative Research Group (1993) A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington's disease chromosomes. Cell 72:971–983. [DOI] [PubMed] [Google Scholar]
 - 300. Thierry AM, Gioanni Y, Degenetais E, Glowinski J (2000) Hippocampo‐prefrontal cortex pathway: anatomical and electrophysiological characteristics. Hippocampus 10:411–419. [DOI] [PubMed] [Google Scholar]
 - 301. Threlfell S, Sammut S, Menniti FS, Schmidt CJ, West AR (2009) Inhibition of phosphodiesterase 10A increases the responsiveness of striatal projection neurons to cortical stimulation. J Pharmacol Exp Ther 328:785–795. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 302. Threlfell S, West AR (2013) Modulation of striatal neuron activity by cyclic nucleotide signaling and phosphodiesterase inhibition. Basal Ganglia 3:137–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 303. Troy CM, Friedman JE, Friedman WJ (2002) Mechanisms of p75‐mediated death of hippocampal neurons. Role of caspases. J Biol Chem 277:34295–34302. [DOI] [PubMed] [Google Scholar]
 - 304. Trueman RC, Brooks SP, Jones L, Dunnett SB (2007) The operant serial implicit learning task reveals early onset motor learning deficits in the Hdh knock‐in mouse model of Huntington's disease. Eur J Neurosci 25:551–558. [DOI] [PubMed] [Google Scholar]
 - 305. Turmaine M, Raza A, Mahal A, Mangiarini L, Bates GP, Davies SW (2000) Nonapoptotic neurodegeneration in a transgenic mouse model of Huntington's disease. Proc Natl Acad Sci USA 97:8093–8097. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 306. Tyebji S, Saavedra A, Canas PM, Pliassova A, Delgado‐Garcia JM, Alberch J et al (2015) Hyperactivation of D1 and A2A receptors contributes to cognitive dysfunction in Huntington's disease. Neurobiol Dis 74:41–57. [DOI] [PubMed] [Google Scholar]
 - 307. Unschuld PG, Joel SE, Liu X, Shanahan M, Margolis RL, Biglan KM et al (2012) Impaired cortico‐striatal functional connectivity in prodromal Huntington's Disease. Neurosci Lett 514:204–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 308. Valor LM, Pulopulos MM, Jimenez‐Minchan M, Olivares R, Lutz B, Barco A (2011) Ablation of CBP in forebrain principal neurons causes modest memory and transcriptional defects and a dramatic reduction of histone acetylation but does not affect cell viability. J Neurosci 31:1652–1663. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 309. van der Zee EA (2014) Synapses, spines and kinases in mammalian learning and memory, and the impact of aging. Neurosci Biobehav Rev 50:77–85. [DOI] [PubMed] [Google Scholar]
 - 310. van Raamsdonk JM, Murphy Z, Slow EJ, Leavitt BR, Hayden MR (2005) Selective degeneration and nuclear localization of mutant huntingtin in the YAC128 mouse model of Huntington disease. Hum Mol Genet 14:3823–3835. [DOI] [PubMed] [Google Scholar]
 - 311. van Raamsdonk JM, Pearson J, Murphy Z, Hayden MR, Leavitt BR (2006) Wild‐type huntingtin ameliorates striatal neuronal atrophy but does not prevent other abnormalities in the YAC128 mouse model of Huntington disease. BMC Neurosci 7:80. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 312. van Raamsdonk JM, Pearson J, Rogers DA, Bissada N, Vogl AW, Hayden MR, Leavitt BR (2005) Loss of wild‐type huntingtin influences motor dysfunction and survival in the YAC128 mouse model of Huntington disease. Hum Mol Genet 14:1379–1392. [DOI] [PubMed] [Google Scholar]
 - 313. van Raamsdonk JM, Pearson J, Slow EJ, Hossain SM, Leavitt BR, Hayden MR (2005) Cognitive dysfunction precedes neuropathology and motor abnormalities in the YAC128 mouse model of Huntington's disease. J Neurosci 25:4169–4180. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 314. van Raamsdonk JM, Warby SC, Hayden MR (2007) Selective degeneration in YAC mouse models of Huntington disease. Brain Res Bull 72:124–131. [DOI] [PubMed] [Google Scholar]
 - 315. Vecsey CG, Hawk JD, Lattal KM, Stein JM, Fabian SA, Attner MA et al (2007) Histone deacetylase inhibitors enhance memory and synaptic plasticity via CREB:CBP‐dependent transcriptional activation. J Neurosci 27:6128–6140. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 316. Verny C, Allain P, Prudean A, Malinge MC, Gohier B, Scherer C et al (2007) Cognitive changes in asymptomatic carriers of the Huntington disease mutation gene. Eur J Neurol 14:1344–1350. [DOI] [PubMed] [Google Scholar]
 - 317. Victorson D, Carlozzi NE, Frank S, Beaumont JL, Cheng W, Gorin B et al (2014) Identifying motor, emotional‐behavioral, and cognitive deficits that comprise the triad of HD symptoms from patient, caregiver, and provider perspectives. Tremor Other Hyperkinet Mov (NY) 4:224. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 318. Vo N, Goodman RH (2001) CREB‐binding protein and p300 in transcriptional regulation. J Biol Chem 276:13505–13508. [DOI] [PubMed] [Google Scholar]
 - 319. Voermans NC, Petersson KM, Daudey L, Weber B, Van Spaendonck KP, Kremer HP, Fernandez G (2004) Interaction between the human hippocampus and the caudate nucleus during route recognition. Neuron 43:427–435. [DOI] [PubMed] [Google Scholar]
 - 320. von Bohlen Und HO (2009) Structure and function of dendritic spines within the hippocampus. Ann Anat 191:518–531. [DOI] [PubMed] [Google Scholar]
 - 321. Vonsattel JP, DiFiglia M (1998) Huntington disease. J Neuropathol Exp Neurol 57:369–384. [DOI] [PubMed] [Google Scholar]
 - 322. Vonsattel JP, Myers RH, Stevens TJ, Ferrante RJ, Bird ED, Richardson EP Jr. (1985) Neuropathological classification of Huntington's disease. J Neuropathol Exp Neurol 44:559–577. [DOI] [PubMed] [Google Scholar]
 - 323. Wang N, Gray M, Lu XH, Cantle JP, Holley SM, Greiner E et al (2014) Neuronal targets for reducing mutant huntingtin expression to ameliorate disease in a mouse model of Huntington's disease. Nat Med 20:536–541. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 324. Wheeler VC, Auerbach W, White JK, Srinidhi J, Auerbach A, Ryan A et al (1999) Length‐dependent gametic CAG repeat instability in the Huntington's disease knock‐in mouse. Hum Mol Genet 8:115–122. [DOI] [PubMed] [Google Scholar]
 - 325. Wheeler VC, Gutekunst CA, Vrbanac V, Lebel LA, Schilling G, Hersch S et al (2002) Early phenotypes that presage late‐onset neurodegenerative disease allow testing of modifiers in Hdh CAG knock‐in mice. Hum Mol Genet 11:633–640. [DOI] [PubMed] [Google Scholar]
 - 326. Wheeler VC, Lebel LA, Vrbanac V, Teed A, te RH, MacDonald ME (2003) Mismatch repair gene Msh2 modifies the timing of early disease in Hdh(Q111) striatum. Hum Mol Genet 12:273–281. [DOI] [PubMed] [Google Scholar]
 - 327. Wheeler VC, White JK, Gutekunst CA, Vrbanac V, Weaver M, Li XJ et al (2000) Long glutamine tracts cause nuclear localization of a novel form of huntingtin in medium spiny striatal neurons in HdhQ92 and HdhQ111 knock‐in mice. Hum Mol Genet 9:503–513. [DOI] [PubMed] [Google Scholar]
 - 328. Wong HK, Liu XB, Matos MF, Chan SF, Perez‐Otano I, Boysen M et al (2002) Temporal and regional expression of NMDA receptor subunit NR3A in the mammalian brain. J Comp Neurol 450:303–317. [DOI] [PubMed] [Google Scholar]
 - 329. Woo NH, Teng HK, Siao CJ, Chiaruttini C, Pang PT, Milner TA et al (2005) Activation of p75NTR by proBDNF facilitates hippocampal long‐term depression. Nat Neurosci 8:1069–1077. [DOI] [PubMed] [Google Scholar]
 - 330. Wood MA, Kaplan MP, Park A, Blanchard EJ, Oliveira AM, Lombardi TL, Abel T (2005) Transgenic mice expressing a truncated form of CREB‐binding protein (CBP) exhibit deficits in hippocampal synaptic plasticity and memory storage. Learn Mem 12:111–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 331. Wu J, Ryskamp DA, Liang X, Egorova P, Zakharova O, Hung G, Bezprozvanny I (2016) Enhanced store‐operated calcium entry leads to striatal synaptic loss in a Huntington's disease mouse model. J Neurosci 36:125–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 332. Wu J, Shih HP, Vigont V, Hrdlicka L, Diggins L, Singh C et al (2011) Neuronal store‐operated calcium entry pathway as a novel therapeutic target for Huntington’ disease treatment. Chem Biol 18:777–793. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 333. Wu ZL, O'Kane TM, Scott RW, Savage MJ, Bozyczko‐Coyne D (2002) Protein tyrosine phosphatases are up‐regulated and participate in cell death induced by polyglutamine expansion. J Biol Chem 277:44208–44213. [DOI] [PubMed] [Google Scholar]
 - 334. Xie Y, Hayden MR, Xu B (2010) BDNF overexpression in the forebrain rescues Huntington's disease phenotypes in YAC128 mice. J Neurosci 30:14708–14718. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 335. Xie Z, Cahill ME, Radulovic J, Wang J, Campbell SL, Miller CA et al (2011) Hippocampal phenotypes in kalirin‐deficient mice. Mol Cell Neurosci 46:45–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 336. Xifro X, Anglada‐Huguet M, Rue L, Saavedra A, Perez‐Navarro E, Alberch J (2011) Increased 90‐kDa ribosomal S6 kinase (Rsk) activity is protective against mutant huntingtin toxicity. Mol Neurodegener 6:74. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 337. Xifro X, Garcia‐Martinez JM, del TD, Alberch J, Perez‐Navarro E (2008) Calcineurin is involved in the early activation of NMDA‐mediated cell death in mutant huntingtin knock‐in striatal cells. J Neurochem 105:1596–1612. [DOI] [PubMed] [Google Scholar]
 - 338. Xifro X, Giralt A, Saavedra A, Garcia‐Martinez JM, Diaz‐Hernandez M, Lucas JJ et al, (2009) Reduced calcineurin protein levels and activity in exon‐1 mouse models of Huntington's disease: role in excitotoxicity. Neurobiol Dis 36:461–469. [DOI] [PubMed] [Google Scholar]
 - 339. Xu Y, Zhang HT, O'Donnell JM (2011) Phosphodiesterases in the central nervous system: implications in mood and cognitive disorders. Handb Exp Pharmacol 204:447–485. [DOI] [PubMed] [Google Scholar]
 - 340. Yamamoto A, Lucas JJ, Hen R (2000) Reversal of neuropathology and motor dysfunction in a conditional model of Huntington's disease. Cell 101:57–66. [DOI] [PubMed] [Google Scholar]
 - 341. Yamashita T, Tucker KL, Barde YA (1999) Neurotrophin binding to the p75 receptor modulates Rho activity and axonal outgrowth. Neuron 24:585–593. [DOI] [PubMed] [Google Scholar]
 - 342. Yhnell E, Dunnett SB, Brooks SP (2016) A longitudinal motor characterisation of the hdhq111 mouse model of Huntington’ disease. J Huntingtons Dis 5:149–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 343. Yhnell E, Lelos MJ, Dunnett SB, Brooks SP (2016) Cognitive training modifies disease symptoms in a mouse model of Huntington's disease. Exp Neurol 282:19–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 344. Yu ZX, Li SH, Evans J, Pillarisetti A, Li H, Li XJ (2003) Mutant huntingtin causes context‐dependent neurodegeneration in mice with Huntington's disease. J Neurosci 23:2193–2202. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 345. Zagrebelsky M, Holz A, Dechant G, Barde YA, Bonhoeffer T, Korte M (2005) The p75 neurotrophin receptor negatively modulates dendrite complexity and spine density in hippocampal neurons. J Neurosci 25:9989–9999. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 346. Zakzanis KK (1998) The subcortical dementia of Huntington's disease. J Clin Exp Neuropsychol 20:565–578. [DOI] [PubMed] [Google Scholar]
 - 347. Zhang H, Wu L, Pchitskaya E, Zakharova O, Saito T, Saido T, Bezprozvanny I (2015) Neuronal store‐operated calcium entry and mushroom spine loss in amyloid precursor protein knock‐in mouse model of Alzheimer's disease. J Neurosci 35:13275–13286. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 348. Zuccato C, Cattaneo E (2007) Role of brain‐derived neurotrophic factor in Huntington’ disease. Prog Neurobiol 81:294–330. [DOI] [PubMed] [Google Scholar]
 - 349. Zuccato C, Cattaneo E (2009) Brain‐derived neurotrophic factor in neurodegenerative diseases. Nat Rev Neurol 5:311–322. [DOI] [PubMed] [Google Scholar]
 - 350. Zuccato C, Ciammola A, Rigamonti D, Leavitt BR, Goffredo D, Conti L et al (2001) Loss of huntingtin‐mediated BDNF gene transcription in Huntington’ disease. Science 293:493–498. [DOI] [PubMed] [Google Scholar]
 - 351. Zuccato C, Liber D, Ramos C, Tarditi A, Rigamonti D, Tartari M et al (2005) Progressive loss of BDNF in a mouse model of Huntington's disease and rescue by BDNF delivery. Pharmacol Res 52:133–139. [DOI] [PubMed] [Google Scholar]
 - 352. Zuccato C, Marullo M, Conforti P, MacDonald ME, Tartari M, Cattaneo E (2008) Systematic assessment of BDNF and its receptor levels in human cortices affected by Huntington’ disease. Brain Pathol 18:225–238. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 353. Zuccato C, Tartari M, Crotti A, Goffredo D, Valenza M, Conti L et al (2003) Huntingtin interacts with REST/NRSF to modulate the transcription of NRSE‐controlled neuronal genes. Nat Genet 35:76–83. [DOI] [PubMed] [Google Scholar]
 - 354. Zuccato C, Valenza M, Cattaneo E (2010) Molecular mechanisms and potential therapeutical targets in Huntington's disease. Physiol Rev 90:905–981. [DOI] [PubMed] [Google Scholar]
 
