Skip to main content
Erschienen in: Molecular Brain 1/2018

Open Access 01.12.2018 | Research

Inhibition of DREAM-ATF6 interaction delays onset of cognition deficit in a mouse model of Huntington’s disease

verfasst von: Alejandro López-Hurtado, Daniel F. Burgos, Paz González, Xose M. Dopazo, Valentina González, Alberto Rábano, Britt Mellström, Jose R. Naranjo

Erschienen in: Molecular Brain | Ausgabe 1/2018

Abstract

The transcriptional repressor DREAM (downstream regulatory element antagonist modulator) is a multifunctional neuronal calcium sensor (NCS) that controls Ca2+ and protein homeostasis through gene regulation and protein-protein interactions. Downregulation of DREAM is part of an endogenous neuroprotective mechanism that improves ATF6 (activating transcription factor 6) processing, neuronal survival in the striatum, and motor coordination in R6/2 mice, a model of Huntington’s disease (HD). Whether modulation of DREAM activity can also ameliorate cognition deficits in HD mice has not been studied. Moreover, it is not known whether DREAM downregulation in HD is unique, or also occurs for other NCS family members. Using the novel object recognition test, we show that chronic administration of the DREAM-binding molecule repaglinide, or induced DREAM haplodeficiency delays onset of cognitive impairment in R6/1 mice, another HD model. The mechanism involves a notable rise in the levels of transcriptionally active ATF6 protein in the hippocampus after repaglinide administration. In addition, we show that reduction in DREAM protein in the hippocampus of HD patients was not accompanied by downregulation of other NCS family members. Our results indicate that DREAM inhibition markedly improves ATF6 processing in the hippocampus and that it might contribute to a delay in memory decline in HD mice. The mechanism of neuroprotection through DREAM silencing in HD does not apply to other NCS family members.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s13041-018-0359-6) contains supplementary material, which is available to authorized users.
Abkürzungen
AD
Alzheimer’s disease
ATF6
Activating transcription factor 6
DREAM
Downstream antagonist modulator
HD
Huntington’s disease
KChIP
Potassium channel interactive protein
LTD
Long-term depression
LTP
Long-term potentiation
mHtt
Mutated huntingtin protein
NCS
Neuronal calcium sensor
NCS-1
Neuronal calcium sensor 1
UPR
Unfolded protein response
VILIP-1
Visinin-like protein 1

Introduction

Huntington’s disease is a devastating, dominantly inherited neurodegenerative disorder caused by expansion of the number of CAG triplets in the first exon of the huntingtin (htt) gene. Expression of mutated Htt (mHtt) induces profound changes in calcium and protein homeostasis that lead ultimately to transcriptional deregulation and synaptic dysfunction [1]. Synaptic dysfunction in HD, and in other neurodegenerative diseases including Alzheimer’s and Parkinson’s diseases, is largely related to failed protein homeostasis because of a defective unfolded protein response (UPR) and accumulation of pathogenic protein aggregates at synapses [26]. Defective UPR in HD is associated with reduced ATF6 processing and a poor pro-adaptive UPR response in the striatum of patients and in HD mouse models [7, 8]. HD neuropathology involves the nearly total loss of motor, cognitive, and emotional control, which is associated with selective death of striatal medium spiny neurons as well as of cortical neurons, the majority of which project to the striatum [1].
DREAM, also known as calsenilin or KChIP3, is a NCS protein that regulates Ca2+ homeostasis and neuronal survival through transcriptional control of target genes and through protein-protein interactions [912]. Transcriptomic and ChIP-seq analyses showed that DREAM is a master-switch transcription factor that regulates the on/off status of specific Ca2+-dependent gene expression programs that control synaptic plasticity, learning, and memory [13, 14]. Long-term depression (LTD), a form of synaptic plasticity, contextual fear, and spatial memory, as well as behavioral anxiety are impaired in transgenic mice that overexpress a dominant active mutant of DREAM (daDREAM) [15]. DREAM-deficient mice show changes in fear conditioning tests [16, 17] and a slight increase in long-term potentiation (LTP) in the dentate gyrus of the hippocampal formation [18]. The mechanism involves postsynaptic modulation of NMDA (N-methyl-D-aspartic acid) receptors by DREAM through a Ca2+-dependent interaction with PSD-95 (post-synaptic density protein 95) [15], or by direct interaction with the NMDA-R1 subunit [19]. In addition, neuronal expression of daDREAM in daDREAM transgenic mice resulted in a complex phenotype that shows i) loss of recurrent inhibition and enhanced LTP in the dentate gyrus as well as impaired learning and memory [13], ii) changes in the expression of specific activity-dependent transcription factors in the hippocampus, including Npas4, Nr4a1 and c-Fos; in addition, these mice have iii) changes in the expression of genes related to the cytoskeleton such as Arc, formin 1 and gelsolin, which are responsible for specific changes in dendritic arborization and spine density in CA1 pyramidal neurons and granule cells of the dentate gyrus [14]. Together these changes recapitulate the role of DREAM in structural plasticity in the hippocampus.
DREAM expression is reduced in murine HD models and in HD patients compared to wild-type mice or healthy individuals [8]. In the R6/2 HD mouse model, decreased DREAM levels are detectable already a few weeks after birth, well before the onset of disease symptoms. Genetic experiments indicated that DREAM downregulation is part of an endogenous neuroprotective mechanism to counteract an equally early failure in ATF6 processing and UPR dysfunction [8]. Chronic administration of repaglinide, an oral hypoglycemic drug that interacts with DREAM, enhances ATF6 processing, which improves the UPR pro-survival function and reduces neuronal loss in the R6/2 mouse striatum [8]. The mechanism involves repaglinide disruption of the Ca2+-dependent DREAM-ATF6 interaction and nuclear accumulation of transcriptionally active ATF6. Improved UPR after chronic repaglinide administration results in delayed onset and slowed progression of motor disease symptoms [8].
Reduced DREAM mRNA levels are also observed in the hippocampus and the cerebral cortex in HD mouse models [8], and decreased nuclear ATF6 immunoreactivity was reported in cortical neurons from HD patients [7]. It is currently not known whether these observations are indicative of an equivalent neuroprotective mechanism by DREAM downregulation that prevents cognitive decline in HD. Here we show that blockade of DREAM activity by repaglinide or by induced DREAM haplodeficiency delayed onset of memory deficits in adult R6/1 mice. We also show that DREAM expression is reduced in the hippocampus of HD patients, while there is no change for other NCS proteins.

Methods

Mice and in vivo treatment

R6/1 mice were originally obtained from Jackson Laboratories. The colony was maintained by breeding male R6/1 with CBA × C57BL/6 mice to obtain heterozygous mutants and wild-type offspring. Genotype and CAG-repeat length were determined by PCR-based amplification using genomic DNA extracted from tail biopsies. Our R6/1 colony had an average repeat length close to 150 repeats, more than the110 repeats originally reported for this mutant transgenic line [20]. Only R6/1 mice with fewer than 170 CAG repeats were used in these experiments. Repaglinide (2 μg/ml) or vehicle (DMSO; 0.2 μl/ml) was administered chronically in drinking water shortly after weaning.

Behavioral analysis

Experiments were performed in R6/1 mice and wild-type littermates of the indicated ages. Mice were initially housed five per cage in a temperature- (21 ± 1 °C) and humidity- (65 ± 10%) controlled room with a 12/12-h light/dark cycle (lights on from 08:00 to 20:00 h), with food and water ad libitum. All experiments took place during the light phase. All behavioral experiments were carried out in blind conditions for genotype and treatment.
The rotarod test was used to measure motor coordination and balance (Accelerating Model, Ugo Basile, Biological Research Apparatus). For basal rotarod performance, mice were tested on two consecutive days. On day 1 (training), each mouse was placed on the rotarod at a constant speed (4 r.p.m.) for a maximum of 60 s. The procedure was repeated three times with a rest period of 30 min between trials. On day 2 (experiment), mice received one training trial at constant speed (4 r.p.m.) for a maximum of 60 s, followed by three test trials with acceleration from 4 to 40 r.p.m. over a period of 60 s and the latency to fall off the rotarod within this period was recorded. Any mice remaining on the apparatus after 60 s were removed and their time scored as 60 s. Data from the three test trials were averaged for each animal and used for statistical analyses.
The novel object recognition test was performed as reported [21, 22]. In brief, mice were first individually habituated to the open-field for 50 min. The next day, they were submitted to a 10-min acquisition trial (first trial) during which they were placed in the open-field in the presence of object A. The time the animal took to explore object A (animal’s snout directed toward the object at a distance < 1 cm) was recorded. Two 10-min retention trials occurred 4 h later (second trial) and 24 h later (third trial). During the second and third trials, objects A and B (second trial) or C (third trial) were placed in the open-field, and the times the animal took to explore object A (tA) and the novel objects B or C (tN) were recorded. A discrimination index was defined as [(tN – tA)/(tA + tN)] × 100. Mice that explored less than five seconds during the initial ten minutes acquisition trial were excluded from the test. Similarly, mice exploring less than five seconds during a given retention trial (4 or 24 h) were excluded from that analysis.

Western blot

Hippocampal whole cell extracts were prepared as described [14]. In brief, brain tissue was homogenized on ice in RIPA buffer (9806, Cell Signaling Technology) supplemented with protease inhibitor (cOmplete EDTA-free, Roche) and 1 mM PMSF (phenylmethanesulfonyl fluoride). Extracts were cleared by centrifugation (14,000 g, 20 min. 4 °C). Samples (20–30 μg) were analyzed by SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis) and immunoblot. PVDF (polyvinylidene difluoride) membranes were incubated (overnight, 4 °C) with specific antibodies to DREAM (Ab731, [23]), ATF6α (A303–719, Bethyl), hippocalcin (G-8, Santa Cruz), VILIP-1 (C-term, Abgent) or to NCS-1 (C-15, Santa Cruz). Equal protein loading was measured by Coomassie staining of the membrane after immunoblotting (Additional file 1). Secondary antibodies used were HRP (horseradish peroxidase)-conjugated donkey anti-rabbit, −mouse or -goat IgG antibody (Jackson) and detection was with ECL Select (GE Healthcare). Lane and band intensity were quantified with ImageLab software (BioRad).

Statistical analyses

All data values are shown as mean ± SEM. Differences were considered significant at P < 0.05. When possible, two-way ANOVA was used to analyze statistical differences among groups. In the case of unequal or small sample size or non-Gaussian distribution, comparisons between groups were analyzed using the nonparametric ANOVA, Kruskal-Wallis test with Dunn’s multiple comparisons between groups. Two-group comparison was performed with unpaired 2-tailed Student’s t test. Animal experiments were randomized. Sample size was not predetermined by statistical method. Prism GraphPad Software 6.0 was used to plot graphs and for statistical analysis.

Results

Chronic repaglinide administration delays onset of memory impairment in R6/1 mice

Using the novel object recognition test, we found that short- and long-term memory were significantly impaired at early stages of the Huntington’s pathology in R6/1 mice. These data confirmed previous reports [2426]. The cognitive decline was already noticeable at 16 weeks after birth (Fig. 1a). Chronic repaglinide administration (2 μg/ml; ad lib in drinking water), which was begun shortly after weaning, partially prevented this decline and restored discrimination ability at 4 h after the test (short-term memory). Repaglinide nonetheless did not improve cognition decline when tested 24 h later (long-term memory) (Fig. 1a). Impaired short- and long-term memory were also observed in 20-week-old R6/1 mice, although chronic repaglinide administration had no effect on cognition loss at this disease stage (Fig. 1b); repaglinide nonetheless effectively reduced the post-prandial increase in circulating glucose levels in 20-week-old R6/1 mice (Fig. 1c). As reported in R6/2 mice [8], extended repaglinide administration in R6/1 or wild-type littermates produced no obvious adverse effects and had no effect on progressive body weight loss in transgenic R6/1 mice (Fig. 1d).
Parallel assessment of R6/1 mice in the rotarod test showed impaired motor coordination at 16 weeks, which became more pronounced by 20 weeks after birth (Fig. 2), as reported [2426]. Chronic repaglinide administration blocked motor dysfunction at 16 weeks, but had no effect in 20-week-old R6/1 mice (Fig. 2). The transient effect of repaglinide in R6/1 mice is similar to its lack of effect on motor coordination in R6/2 mice at more advanced stages of the disease [8].
Ablation of one copy of DREAM in double transgenic R6/1xDREAM+/− mice significantly improved the discrimination index at 4 and 24 h after exposure to the novel objects in 16-week-old mice, whereas the effect was not significant in 20-week-old mice (Fig. 3). These results parallel the delay of the onset of motor symptoms in R6/2 mice after induced DREAM haplodeficiency [8].

Repaglinide administration normalizes ATF6 processing in the R6/1 mouse hippocampus

Reduced DREAM mRNA expression and protein levels in the brain of R6/1 and R6/2 mice is a neuroprotective response, and further inhibition of DREAM activity is associated with improved motor coordination in the HD mice treated with repaglinide [8]. To test whether a similar mechanism is involved in the partial protection from cognitive decline in repaglinide-treated HD mice, we analyzed the effect of repaglinide administration on ATF6 processing in R6/1 hippocampus, a brain area involved in learning and memory [27].
ATF6 processing, measured as the ratio between the processed (p50) and the full-length (p100) forms of the protein, was markedly reduced in the R6/1 mouse hippocampus compared to wild-type littermates (Fig. 4). Repaglinide normalized ATF6 processing in the R6/1 mouse hippocampus without changing ATF6 processing in wild-type littermates (Fig. 4). Recovery of ATF6 processing in the R6/1 hippocampus thus correlates with delayed cognition impairment after repaglinide administration.

Differential changes in NCS protein expression in the hippocampus of HD patients

Repaglinide binds to DREAM and to other members of the NCS superfamily [8, 28], and changes in expression of some NCS proteins have been described in HD patients [29]. We therefore analyzed whether expression of three NCS proteins other than DREAM is also reduced in hippocampus from HD patients, and might be targeted by repaglinide.
As shown in the striatum from HD patients [8], DREAM protein levels are notably reduced in hippocampal samples from HD patients compared to control samples (Fig. 5). Neuronal calcium sensor-1 (NCS-1), visinin-like protein (VILIP-1) and hippocalcin levels were nonetheless similar in the same control and HD hippocampus samples (Fig. 5). These results further contribute to the idea that reduced DREAM levels in the hippocampus of HD patients may have a neuroprotective effect to limit and/or slow down cognitive decline in HD.

Discussion

Early synaptic dysfunction and progressive accumulation of pathogenic protein aggregates (e.g., huntingtin inclusions, amyloid plaques, and neurofibrillary tangles) lead to gradual, inescapable cognitive impairment and neuronal death in HD and Alzheimer’s disease (AD). In the case of HD, early symptoms of synaptic dysfunction in the cortico-striatal pathway include changes in NMDA receptor signaling, reduced response to external stimuli (e.g., impaired induction of activity-dependent genes), progressive loss of synaptic contacts (e.g., post-synaptic dendritic spines in excitatory synapses), and gradual degeneration of medium-sized striatal spiny neurons [30]. Reduction in hippocampal volume is reported in HD patients [31, 32], which correlates with altered spatial short-term and recognition memories in these patients [33, 34]. Cognitive decline precedes motor manifestations, both in patients and in HD mouse models [3537].
The NCS superfamily is encoded by 14 genes in mammals and, through alternative splicing, encompasses more than 40 isoforms [38, 39]. Guided by multiple Ca2+-dependent and -independent protein-protein interactions, and by a specific Ca2+-dependent interaction with DNA in DREAM/KChIP subfamily members [9, 40], NCS proteins participate in numerous physiological functions [38, 39]. In addition, changes in the expression of or mutations in NCS proteins are associated with several neurological disorders. NCS-1 is upregulated in patients with schizophrenia or bipolar disorder [41] as well as in the substantia nigra from Parkinson’s disease patients [42], and a mutation in NCS-1 was found in a case of autistic spectrum disorder [43, 44]. Expression of neurocalcin [45] and VILIP-1 [46] were reduced in AD brains compared with age-matched brain samples. Increased VILIP-1 levels in cerebrospinal fluid were proposed as diagnostic and prognostic biomarkers of neuroinflammation and cognitive decline in patients with AD, dementia with Lewy bodies, or frontotemporal lobar degeneration [4753]. Finally, decreased DREAM mRNA and protein levels [8] and hippocalcin mRNA expression [29] were found in striatum from Huntington’s disease patients. This reduction in hippocalcin mRNA nonetheless did not correlate with striatal vulnerability, and the study did not analyze hippocalcin protein levels. Our results show that, of the NCS proteins tested, only DREAM levels were decreased in hippocampal samples from HD patients. These results further extend DREAM involvement in HD, and suggest that NCS-1, hippocalcin, and VILIP-1 are not implicated functionally in this neurodegenerative pathology.
Early downregulation of DREAM expression in the HD mouse striatum is associated with an ATF6-mediated neuroprotective mechanism that delays onset and slows progression of the motor symptoms of the disease [8]. Here we show that DREAM and transcriptionally active ATF6 are also reduced in the hippocampus and that chronic administration of repaglinide improves ATF6 processing and delays memory impairment in adult R6/1 mice. Removal of one copy of DREAM in R6/1xDREAM+/− double transgenic mice similarly delayed onset of cognitive loss, as assayed in the novel object recognition test. As shown for improved motor coordination in R6/2 mice [8], pharmacologically or genetically induced reduction of DREAM activity in R6/1 mice also has a transient effect at the hippocampal level, as cognitive loss was delayed but not prevented in advanced disease stages. The molecular basis for this finding is presently unknown, although it might be related to the activation of additional signaling pathways that lead to irreversible neuronal death and loss of cognitive function. Reduction of mHtt mRNA levels using sustained, neuron-specific expression of synthetic zinc finger constructs that target the CAG repeats in the mHtt gene also provided only transient protection for neuronal loss in R6/1 mice [54].
Repaglinide binding to NCS proteins was first reported in bovine brain and retinal extracts, which showed Ca2+-dependent binding respectively to neurocalcin and VILIP-1, or to recoverin [28]. Repaglinide also binds to members of the DREAM/KChIP subfamily [8], which indicates that repaglinide binding is a characteristic of all proteins of the NCS superfamily. After binding, repaglinide interferes with the biological activity of the Ca2+ sensor, that is, it blocks recoverin-mediated inhibition of rhodopsin kinase activity or DREAM-induced suppression of ATF6 processing [8, 28].
Repaglinide was developed as a potent insulinotropic agent for treatment of type-2 diabetes [55]. The mechanism involves the blockade of ATP-dependent potassium channels, which induces insulin release. Like glibenclamide, another insulinotropic molecule, nanomolar concentrations of repaglinide block these channels. Repaglinide binds to DREAM and blocks activation of Kv4 potassium channels, also at nanomolar concentrations, whereas glibenclamide is inactive [8]. The interaction of repaglinide with NCS proteins is specific for this group, and does not occur with other Ca2+-binding proteins, including calmodulin or proteins of the S-100 superfamily [28].
Our results suggest that DREAM inhibition has a role in delaying cognitive decline in HD mice through a mechanism related to ATF6 processing. This neuroprotection through DREAM silencing in HD is specific, and does not apply to other NCS family members. Ongoing studies that include molecular docking and structure-activity relationship analysis will help to better understand this interaction and, ideally, to define new ligands with improved selectivity for the different NCS subfamilies.

Acknowledgements

We thank C. Mark for editorial assistance.

Funding

This work was funded by the Instituto de Salud Carlos III/CIBERNED (to JRN, BM and AR), the Madrid regional government/Neurodegmodels (to JRN), and SAF2014–53412-R and SAF2017–89554-R (AEI-FEDER, EU) (to JRN).

Availability of data and materials

All data generated or analyzed during this study are included in this published article [and its supplementary information files]. Non-commercial materials used in this study are available from the corresponding author on reasonable request.

Ethics approval

Behavioral tests and animal care were conducted in accordance with standard ethical guidelines (European Communities Directive 86/609 EEC; National Institutes of Health 1995). The CNB-CSIC and Community of Madrid ethical committees approved experiments with mice and use of human post-mortem samples (PROEX 28/05).
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

e.Med Neurologie

Kombi-Abonnement

Mit e.Med Neurologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes, den Premium-Inhalten der neurologischen Fachzeitschriften, inklusive einer gedruckten Neurologie-Zeitschrift Ihrer Wahl.

Literatur
2.
Zurück zum Zitat Lee J, Ozcan U. Unfolded protein response signaling and metabolic diseases. J Biol Chem. 2014;289(3):1203–11.CrossRefPubMed Lee J, Ozcan U. Unfolded protein response signaling and metabolic diseases. J Biol Chem. 2014;289(3):1203–11.CrossRefPubMed
3.
Zurück zum Zitat Halliday M, Mallucci GR. Modulating the unfolded protein response to prevent neurodegeneration and enhance memory. Neuropathol Appl Neurobiol. 2015;41(4):414–27.CrossRefPubMedPubMedCentral Halliday M, Mallucci GR. Modulating the unfolded protein response to prevent neurodegeneration and enhance memory. Neuropathol Appl Neurobiol. 2015;41(4):414–27.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Hetz C, Mollereau B. Disturbance of endoplasmic reticulum proteostasis in neurodegenerative diseases. Nat Rev Neurosci. 2014;15(4):233–49.CrossRefPubMed Hetz C, Mollereau B. Disturbance of endoplasmic reticulum proteostasis in neurodegenerative diseases. Nat Rev Neurosci. 2014;15(4):233–49.CrossRefPubMed
5.
Zurück zum Zitat Sprenkle NT, Sims SG, Sanchez CL, Meares GP. Endoplasmic reticulum stress and inflammation in the central nervous system. Mol Neurodegener. 2017;12(1):42.CrossRefPubMedPubMedCentral Sprenkle NT, Sims SG, Sanchez CL, Meares GP. Endoplasmic reticulum stress and inflammation in the central nervous system. Mol Neurodegener. 2017;12(1):42.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Herms J, Dorostkar MM. Dendritic spine pathology in neurodegenerative diseases. Annu Rev Pathol. 2016;11:221–50.CrossRefPubMed Herms J, Dorostkar MM. Dendritic spine pathology in neurodegenerative diseases. Annu Rev Pathol. 2016;11:221–50.CrossRefPubMed
7.
Zurück zum Zitat Fernandez-Fernandez MR, Ferrer I, Lucas JJ. Impaired ATF6alpha processing, decreased Rheb and neuronal cell cycle re-entry in Huntington's disease. Neurobiol Dis. 2011;41(1):23–32.CrossRefPubMed Fernandez-Fernandez MR, Ferrer I, Lucas JJ. Impaired ATF6alpha processing, decreased Rheb and neuronal cell cycle re-entry in Huntington's disease. Neurobiol Dis. 2011;41(1):23–32.CrossRefPubMed
8.
Zurück zum Zitat Naranjo JRZH, Villar D, González P, Dopazo XM, Morón J, Higueras E, Oliveros JC, Arrabal MD, Prieto A, Cercós P, González T, De la Cruz A, Casado-Vela J, Rábano A, Valenzuela C, Gutierrez-Rodriguez M, Li JY, Mellström B. Activating transcription factor 6 de-repression mediates neuroprotection in Huntington's disease. J Clin Invest. 2016;126(2):627–38.CrossRefPubMedPubMedCentral Naranjo JRZH, Villar D, González P, Dopazo XM, Morón J, Higueras E, Oliveros JC, Arrabal MD, Prieto A, Cercós P, González T, De la Cruz A, Casado-Vela J, Rábano A, Valenzuela C, Gutierrez-Rodriguez M, Li JY, Mellström B. Activating transcription factor 6 de-repression mediates neuroprotection in Huntington's disease. J Clin Invest. 2016;126(2):627–38.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Carrion AM, Link WA, Ledo F, Mellstrom B, Naranjo JR. DREAM is a Ca2+−regulated transcriptional repressor. Nature. 1999;398(6722):80–4.CrossRefPubMed Carrion AM, Link WA, Ledo F, Mellstrom B, Naranjo JR. DREAM is a Ca2+−regulated transcriptional repressor. Nature. 1999;398(6722):80–4.CrossRefPubMed
10.
Zurück zum Zitat Buxbaum JD, Choi EK, Luo Y, Lilliehook C, Crowley AC, Merriam DE, Wasco W. Calsenilin: a calcium-binding protein that interacts with the presenilins and regulates the levels of a presenilin fragment. Nat Med. 1998;4(10):1177–81.CrossRefPubMed Buxbaum JD, Choi EK, Luo Y, Lilliehook C, Crowley AC, Merriam DE, Wasco W. Calsenilin: a calcium-binding protein that interacts with the presenilins and regulates the levels of a presenilin fragment. Nat Med. 1998;4(10):1177–81.CrossRefPubMed
11.
Zurück zum Zitat An WF, Bowlby MR, Betty M, Cao J, Ling HP, Mendoza G, Hinson JW, Mattsson KI, Strassle BW, Trimmer JS, et al. Modulation of A-type potassium channels by a family of calcium sensors. Nature. 2000;403(6769):553–6.CrossRefPubMed An WF, Bowlby MR, Betty M, Cao J, Ling HP, Mendoza G, Hinson JW, Mattsson KI, Strassle BW, Trimmer JS, et al. Modulation of A-type potassium channels by a family of calcium sensors. Nature. 2000;403(6769):553–6.CrossRefPubMed
12.
Zurück zum Zitat Mellstrom B, Savignac M, Gomez-Villafuertes R, Naranjo JR. Ca2+−operated transcriptional networks: molecular mechanisms and in vivo models. Physiol Rev. 2008;88(2):421–49.CrossRefPubMed Mellstrom B, Savignac M, Gomez-Villafuertes R, Naranjo JR. Ca2+−operated transcriptional networks: molecular mechanisms and in vivo models. Physiol Rev. 2008;88(2):421–49.CrossRefPubMed
13.
Zurück zum Zitat Mellstrom B, Sahun I, Ruiz-Nuno A, Murtra P, Gomez-Villafuertes R, Savignac M, Oliveros JC, Gonzalez P, Kastanauskaite A, Knafo S, et al. DREAM controls the on/off switch of specific activity-dependent transcription pathways. Mol Cell Biol. 2014;34(5):877–87.CrossRefPubMedPubMedCentral Mellstrom B, Sahun I, Ruiz-Nuno A, Murtra P, Gomez-Villafuertes R, Savignac M, Oliveros JC, Gonzalez P, Kastanauskaite A, Knafo S, et al. DREAM controls the on/off switch of specific activity-dependent transcription pathways. Mol Cell Biol. 2014;34(5):877–87.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Mellström BKA, Knafo S, Gonzalez P, Dopazo XM, Ruiz-Nuño A, Jefferys J, Zhuo M, Bliss TVP, Naranjo JR, De Felipe J, Unbalanced DREAM. Activity modifies hippocampal connectivity and cognition. Mol Brain. 2016;9:22–33.CrossRefPubMedPubMedCentral Mellström BKA, Knafo S, Gonzalez P, Dopazo XM, Ruiz-Nuño A, Jefferys J, Zhuo M, Bliss TVP, Naranjo JR, De Felipe J, Unbalanced DREAM. Activity modifies hippocampal connectivity and cognition. Mol Brain. 2016;9:22–33.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Wu LJ, Mellstrom B, Wang H, Ren M, Domingo S, Kim SS, Li XY, Chen T, Naranjo JR, Zhuo M. DREAM (downstream regulatory element antagonist modulator) contributes to synaptic depression and contextual fear memory. Mol Brain. 2010;3:3–15.CrossRefPubMedPubMedCentral Wu LJ, Mellstrom B, Wang H, Ren M, Domingo S, Kim SS, Li XY, Chen T, Naranjo JR, Zhuo M. DREAM (downstream regulatory element antagonist modulator) contributes to synaptic depression and contextual fear memory. Mol Brain. 2010;3:3–15.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Lilliehook C, Bozdagi O, Yao J, Gomez-Ramirez M, Zaidi NF, Wasco W, Gandy S, Santucci AC, Haroutunian V, Huntley GW, et al. Altered Abeta formation and long-term potentiation in a calsenilin knock-out. J Neurosci. 2003;23(27):9097–106.PubMed Lilliehook C, Bozdagi O, Yao J, Gomez-Ramirez M, Zaidi NF, Wasco W, Gandy S, Santucci AC, Haroutunian V, Huntley GW, et al. Altered Abeta formation and long-term potentiation in a calsenilin knock-out. J Neurosci. 2003;23(27):9097–106.PubMed
17.
Zurück zum Zitat Alexander JC, McDermott CM, Tunur T, Rands V, Stelly C, Karhson D, Bowlby MR, An WF, Sweatt JD, Schrader LA. The role of calsenilin/DREAM/KChIP3 in contextual fear conditioning. Learn Mem. 2009;16(3):167–77.CrossRefPubMedPubMedCentral Alexander JC, McDermott CM, Tunur T, Rands V, Stelly C, Karhson D, Bowlby MR, An WF, Sweatt JD, Schrader LA. The role of calsenilin/DREAM/KChIP3 in contextual fear conditioning. Learn Mem. 2009;16(3):167–77.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Cheng HY, Pitcher GM, Laviolette SR, Whishaw IQ, Tong KI, Kockeritz LK, Wada T, Joza NA, Crackower M, Goncalves J, et al. DREAM is a critical transcriptional repressor for pain modulation. Cell. 2002;108(1):31–43.CrossRefPubMed Cheng HY, Pitcher GM, Laviolette SR, Whishaw IQ, Tong KI, Kockeritz LK, Wada T, Joza NA, Crackower M, Goncalves J, et al. DREAM is a critical transcriptional repressor for pain modulation. Cell. 2002;108(1):31–43.CrossRefPubMed
19.
Zurück zum Zitat Zhang Y, Su P, Liang P, Liu T, Liu X, Liu XY, Zhang B, Han T, Zhu YB, Yin DM, et al. The DREAM protein negatively regulates the NMDA receptor through interaction with the NR1 subunit. J Neurosci. 2010;30(22):7575–86.CrossRefPubMed Zhang Y, Su P, Liang P, Liu T, Liu X, Liu XY, Zhang B, Han T, Zhu YB, Yin DM, et al. The DREAM protein negatively regulates the NMDA receptor through interaction with the NR1 subunit. J Neurosci. 2010;30(22):7575–86.CrossRefPubMed
20.
Zurück zum Zitat Mangiarini L, Sathasivam K, Seller M, Cozens B, Harper A, Hetherington C, Lawton M, Trottier Y, Lehrach H, Davies SW, et al. Exon 1 of the HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice. Cell. 1996;87(3):493–506.CrossRefPubMed Mangiarini L, Sathasivam K, Seller M, Cozens B, Harper A, Hetherington C, Lawton M, Trottier Y, Lehrach H, Davies SW, et al. Exon 1 of the HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice. Cell. 1996;87(3):493–506.CrossRefPubMed
21.
Zurück zum Zitat Tan VTY, Mockett BG, Ohline SM, Parfitt KD, Wicky HE, Peppercorn K, Schoderboeck L, Yahaya MFB, Tate WP, Hughes SM, et al. Lentivirus-mediated expression of human secreted amyloid precursor protein-alpha prevents development of memory and plasticity deficits in a mouse model of Alzheimer's disease. Mol Brain. 2018;11(1):7.CrossRefPubMedPubMedCentral Tan VTY, Mockett BG, Ohline SM, Parfitt KD, Wicky HE, Peppercorn K, Schoderboeck L, Yahaya MFB, Tate WP, Hughes SM, et al. Lentivirus-mediated expression of human secreted amyloid precursor protein-alpha prevents development of memory and plasticity deficits in a mouse model of Alzheimer's disease. Mol Brain. 2018;11(1):7.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Cui L, Sun W, Yu M, Li N, Guo L, Gu H, Zhou Y. Disrupted-in-schizophrenia1 (DISC1) L100P mutation alters synaptic transmission and plasticity in the hippocampus and causes recognition memory deficits. Mol Brain. 2016;9(1):89–101.CrossRefPubMedPubMedCentral Cui L, Sun W, Yu M, Li N, Guo L, Gu H, Zhou Y. Disrupted-in-schizophrenia1 (DISC1) L100P mutation alters synaptic transmission and plasticity in the hippocampus and causes recognition memory deficits. Mol Brain. 2016;9(1):89–101.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Savignac M, Pintado B, Gutierrez-Adan A, Palczewska M, Mellstrom B, Naranjo JR. Transcriptional repressor DREAM regulates T-lymphocyte proliferation and cytokine gene expression. EMBO J. 2005;24(20):3555–64.CrossRefPubMedPubMedCentral Savignac M, Pintado B, Gutierrez-Adan A, Palczewska M, Mellstrom B, Naranjo JR. Transcriptional repressor DREAM regulates T-lymphocyte proliferation and cytokine gene expression. EMBO J. 2005;24(20):3555–64.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat van der Borght K, Brundin P. Reduced expression of PSA-NCAM in the hippocampus and piriform cortex of the R6/1 and R6/2 mouse models of Huntington's disease. Exp Neurol. 2007;204(1):473–8.CrossRefPubMed van der Borght K, Brundin P. Reduced expression of PSA-NCAM in the hippocampus and piriform cortex of the R6/1 and R6/2 mouse models of Huntington's disease. Exp Neurol. 2007;204(1):473–8.CrossRefPubMed
25.
Zurück zum Zitat Li W, Silva HB, Real J, Wang YM, Rial D, Li P, Payen MP, Zhou Y, Muller CE, Tome AR, et al. Inactivation of adenosine A2A receptors reverses working memory deficits at early stages of Huntington's disease models. Neurobiol Dis. 2015;79:70–80.CrossRefPubMed Li W, Silva HB, Real J, Wang YM, Rial D, Li P, Payen MP, Zhou Y, Muller CE, Tome AR, et al. Inactivation of adenosine A2A receptors reverses working memory deficits at early stages of Huntington's disease models. Neurobiol Dis. 2015;79:70–80.CrossRefPubMed
26.
Zurück zum Zitat Tyebji S, Saavedra A, Canas PM, Pliassova A, Delgado-Garcia JM, Alberch J, Cunha RA, Gruart A, Perez-Navarro E. Hyperactivation of D1 and A2A receptors contributes to cognitive dysfunction in Huntington's disease. Neurobiol Dis. 2015;74:41–57.CrossRefPubMed Tyebji S, Saavedra A, Canas PM, Pliassova A, Delgado-Garcia JM, Alberch J, Cunha RA, Gruart A, Perez-Navarro E. Hyperactivation of D1 and A2A receptors contributes to cognitive dysfunction in Huntington's disease. Neurobiol Dis. 2015;74:41–57.CrossRefPubMed
27.
Zurück zum Zitat Eichenbaum H. The hippocampus and declarative memory: cognitive mechanisms and neural codes. Behav Brain Res. 2001;127(1–2):199–207.CrossRefPubMed Eichenbaum H. The hippocampus and declarative memory: cognitive mechanisms and neural codes. Behav Brain Res. 2001;127(1–2):199–207.CrossRefPubMed
28.
Zurück zum Zitat Okada M, Takezawa D, Tachibanaki S, Kawamura S, Tokumitsu H, Kobayashi R. Neuronal calcium sensor proteins are direct targets of the insulinotropic agent repaglinide. Biochem J. 2003;375(Pt 1):87–97.CrossRefPubMedPubMedCentral Okada M, Takezawa D, Tachibanaki S, Kawamura S, Tokumitsu H, Kobayashi R. Neuronal calcium sensor proteins are direct targets of the insulinotropic agent repaglinide. Biochem J. 2003;375(Pt 1):87–97.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Rudinskiy N, Kaneko YA, Beesen AA, Gokce O, Regulier E, Deglon N, Luthi-Carter R. Diminished hippocalcin expression in Huntington's disease brain does not account for increased striatal neuron vulnerability as assessed in primary neurons. J Neurochem. 2009;111(2):460–72.CrossRefPubMed Rudinskiy N, Kaneko YA, Beesen AA, Gokce O, Regulier E, Deglon N, Luthi-Carter R. Diminished hippocalcin expression in Huntington's disease brain does not account for increased striatal neuron vulnerability as assessed in primary neurons. J Neurochem. 2009;111(2):460–72.CrossRefPubMed
31.
Zurück zum Zitat Rosas HD, Koroshetz WJ, Chen YI, Skeuse C, Vangel M, Cudkowicz ME, Caplan K, Marek K, Seidman LJ, Makris N, et al. Evidence for more widespread cerebral pathology in early HD: an MRI-based morphometric analysis. Neurology. 2003;60(10):1615–20.CrossRefPubMed Rosas HD, Koroshetz WJ, Chen YI, Skeuse C, Vangel M, Cudkowicz ME, Caplan K, Marek K, Seidman LJ, Makris N, et al. Evidence for more widespread cerebral pathology in early HD: an MRI-based morphometric analysis. Neurology. 2003;60(10):1615–20.CrossRefPubMed
32.
Zurück zum Zitat Ille R, Schafer A, Scharmuller W, Enzinger C, Schoggl H, Kapfhammer HP, Schienle A. Emotion recognition and experience in Huntington disease: a voxel-based morphometry study. J Psychiatry Neurosci. 2011;36(6):383–90.CrossRefPubMedPubMedCentral Ille R, Schafer A, Scharmuller W, Enzinger C, Schoggl H, Kapfhammer HP, Schienle A. Emotion recognition and experience in Huntington disease: a voxel-based morphometry study. J Psychiatry Neurosci. 2011;36(6):383–90.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Lemiere J, Decruyenaere M, Evers-Kiebooms G, Vandenbussche E, Dom R. Cognitive changes in patients with Huntington's disease (HD) and asymptomatic carriers of the HD mutation--a longitudinal follow-up study. J Neurol. 2004;251(8):935–42.CrossRefPubMed Lemiere J, Decruyenaere M, Evers-Kiebooms G, Vandenbussche E, Dom R. Cognitive changes in patients with Huntington's disease (HD) and asymptomatic carriers of the HD mutation--a longitudinal follow-up study. J Neurol. 2004;251(8):935–42.CrossRefPubMed
34.
Zurück zum Zitat Montoya A, Pelletier M, Menear M, Duplessis E, Richer F, Lepage M. Episodic memory impairment in Huntington's disease: a meta-analysis. Neuropsychologia. 2006;44(10):1984–94.CrossRefPubMed Montoya A, Pelletier M, Menear M, Duplessis E, Richer F, Lepage M. Episodic memory impairment in Huntington's disease: a meta-analysis. Neuropsychologia. 2006;44(10):1984–94.CrossRefPubMed
36.
Zurück zum Zitat Stout JC, Paulsen JS, Queller S, Solomon AC, Whitlock KB, Campbell JC, Carlozzi N, Duff K, Beglinger LJ, Langbehn DR, et al. Neurocognitive signs in prodromal Huntington disease. Neuropsychology. 2011;25(1):1–14.CrossRefPubMedPubMedCentral Stout JC, Paulsen JS, Queller S, Solomon AC, Whitlock KB, Campbell JC, Carlozzi N, Duff K, Beglinger LJ, Langbehn DR, et al. Neurocognitive signs in prodromal Huntington disease. Neuropsychology. 2011;25(1):1–14.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Puigdellivol M, Saavedra A, Perez-Navarro E. Cognitive dysfunction in Huntington's disease: mechanisms and therapeutic strategies beyond BDNF. Brain Pathol. 2016;26(6):752–71.CrossRefPubMed Puigdellivol M, Saavedra A, Perez-Navarro E. Cognitive dysfunction in Huntington's disease: mechanisms and therapeutic strategies beyond BDNF. Brain Pathol. 2016;26(6):752–71.CrossRefPubMed
40.
Zurück zum Zitat Link WA, Ledo F, Torres B, Palczewska M, Madsen TM, Savignac M, Albar JP, Mellstrom B, Naranjo JR. Day-night changes in downstream regulatory element antagonist modulator/potassium channel interacting protein activity contribute to circadian gene expression in pineal gland. J Neurosci. 2004;24(23):5346–55.CrossRefPubMed Link WA, Ledo F, Torres B, Palczewska M, Madsen TM, Savignac M, Albar JP, Mellstrom B, Naranjo JR. Day-night changes in downstream regulatory element antagonist modulator/potassium channel interacting protein activity contribute to circadian gene expression in pineal gland. J Neurosci. 2004;24(23):5346–55.CrossRefPubMed
41.
Zurück zum Zitat Koh PO, Undie AS, Kabbani N, Levenson R, Goldman-Rakic PS, Lidow MS. Up-regulation of neuronal calcium sensor-1 (NCS-1) in the prefrontal cortex of schizophrenic and bipolar patients. Proc Natl Acad Sci U S A. 2003;100(1):313–7.CrossRefPubMed Koh PO, Undie AS, Kabbani N, Levenson R, Goldman-Rakic PS, Lidow MS. Up-regulation of neuronal calcium sensor-1 (NCS-1) in the prefrontal cortex of schizophrenic and bipolar patients. Proc Natl Acad Sci U S A. 2003;100(1):313–7.CrossRefPubMed
42.
Zurück zum Zitat Dragicevic E, Poetschke C, Duda J, Schlaudraff F, Lammel S, Schiemann J, Fauler M, Hetzel A, Watanabe M, Lujan R, et al. Cav1.3 channels control D2-autoreceptor responses via NCS-1 in substantia nigra dopamine neurons. Brain. 2014;137(Pt 8):2287–302.CrossRefPubMedPubMedCentral Dragicevic E, Poetschke C, Duda J, Schlaudraff F, Lammel S, Schiemann J, Fauler M, Hetzel A, Watanabe M, Lujan R, et al. Cav1.3 channels control D2-autoreceptor responses via NCS-1 in substantia nigra dopamine neurons. Brain. 2014;137(Pt 8):2287–302.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Piton A, Michaud JL, Peng H, Aradhya S, Gauthier J, Mottron L, Champagne N, Lafreniere RG, Hamdan FF, team SD, et al. Mutations in the calcium-related gene IL1RAPL1 are associated with autism. Hum Mol Genet. 2008;17(24):3965–74.CrossRefPubMed Piton A, Michaud JL, Peng H, Aradhya S, Gauthier J, Mottron L, Champagne N, Lafreniere RG, Hamdan FF, team SD, et al. Mutations in the calcium-related gene IL1RAPL1 are associated with autism. Hum Mol Genet. 2008;17(24):3965–74.CrossRefPubMed
44.
Zurück zum Zitat Handley MT, Lian LY, Haynes LP, Burgoyne RD. Structural and functional deficits in a neuronal calcium sensor-1 mutant identified in a case of autistic spectrum disorder. PLoS One. 2010;5(5):e10534.CrossRefPubMedPubMedCentral Handley MT, Lian LY, Haynes LP, Burgoyne RD. Structural and functional deficits in a neuronal calcium sensor-1 mutant identified in a case of autistic spectrum disorder. PLoS One. 2010;5(5):e10534.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Shimohama S, Chachin M, Taniguchi T, Hidaka H, Kimura J. Changes of neurocalcin, a calcium-binding protein, in the brain of patients with Alzheimer's disease. Brain Res. 1996;716(1–2):233–6.CrossRefPubMed Shimohama S, Chachin M, Taniguchi T, Hidaka H, Kimura J. Changes of neurocalcin, a calcium-binding protein, in the brain of patients with Alzheimer's disease. Brain Res. 1996;716(1–2):233–6.CrossRefPubMed
46.
Zurück zum Zitat Braunewell K, Riederer P, Spilker C, Gundelfinger ED, Bogerts B, Bernstein HG. Abnormal localization of two neuronal calcium sensor proteins, visinin-like proteins (vilips)-1 and −3, in neocortical brain areas of Alzheimer disease patients. Dement Geriatr Cogn Disord. 2001;12(2):110–6.CrossRefPubMed Braunewell K, Riederer P, Spilker C, Gundelfinger ED, Bogerts B, Bernstein HG. Abnormal localization of two neuronal calcium sensor proteins, visinin-like proteins (vilips)-1 and −3, in neocortical brain areas of Alzheimer disease patients. Dement Geriatr Cogn Disord. 2001;12(2):110–6.CrossRefPubMed
47.
Zurück zum Zitat Tarawneh R, D'Angelo G, Macy E, Xiong C, Carter D, Cairns NJ, Fagan AM, Head D, Mintun MA, Ladenson JH, et al. Visinin-like protein-1: diagnostic and prognostic biomarker in Alzheimer disease. Ann Neurol. 2011;70(2):274–85.CrossRefPubMedPubMedCentral Tarawneh R, D'Angelo G, Macy E, Xiong C, Carter D, Cairns NJ, Fagan AM, Head D, Mintun MA, Ladenson JH, et al. Visinin-like protein-1: diagnostic and prognostic biomarker in Alzheimer disease. Ann Neurol. 2011;70(2):274–85.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Tarawneh R, Lee JM, Ladenson JH, Morris JC, Holtzman DM. CSF VILIP-1 predicts rates of cognitive decline in early Alzheimer disease. Neurology. 2012;78(10):709–19.CrossRefPubMedPubMedCentral Tarawneh R, Lee JM, Ladenson JH, Morris JC, Holtzman DM. CSF VILIP-1 predicts rates of cognitive decline in early Alzheimer disease. Neurology. 2012;78(10):709–19.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Luo X, Hou L, Shi H, Zhong X, Zhang Y, Zheng D, Tan Y, Hu G, Mu N, Chan J, et al. CSF levels of the neuronal injury biomarker visinin-like protein-1 in Alzheimer's disease and dementia with Lewy bodies. J Neurochem. 2013;127(5):681–90.CrossRefPubMed Luo X, Hou L, Shi H, Zhong X, Zhang Y, Zheng D, Tan Y, Hu G, Mu N, Chan J, et al. CSF levels of the neuronal injury biomarker visinin-like protein-1 in Alzheimer's disease and dementia with Lewy bodies. J Neurochem. 2013;127(5):681–90.CrossRefPubMed
50.
Zurück zum Zitat Mroczko B, Groblewska M, Zboch M, Muszynski P, Zajkowska A, Borawska R, Szmitkowski M, Kornhuber J, Lewczuk P. Evaluation of visinin-like protein 1 concentrations in the cerebrospinal fluid of patients with mild cognitive impairment as a dynamic biomarker of Alzheimer's disease. J Alzheimers Dis. 2015;43(3):1031–7.PubMed Mroczko B, Groblewska M, Zboch M, Muszynski P, Zajkowska A, Borawska R, Szmitkowski M, Kornhuber J, Lewczuk P. Evaluation of visinin-like protein 1 concentrations in the cerebrospinal fluid of patients with mild cognitive impairment as a dynamic biomarker of Alzheimer's disease. J Alzheimers Dis. 2015;43(3):1031–7.PubMed
51.
Zurück zum Zitat Kirkwood CM, MacDonald ML, Schempf TA, Vatsavayi AV, Ikonomovic MD, Koppel JL, Ding Y, Sun M, Kofler JK, Lopez OL, et al. Altered levels of Visinin-like protein 1 correspond to regional neuronal loss in Alzheimer disease and frontotemporal lobar degeneration. J Neuropathol Exp Neurol. 2016;75(2):175–82.CrossRefPubMed Kirkwood CM, MacDonald ML, Schempf TA, Vatsavayi AV, Ikonomovic MD, Koppel JL, Ding Y, Sun M, Kofler JK, Lopez OL, et al. Altered levels of Visinin-like protein 1 correspond to regional neuronal loss in Alzheimer disease and frontotemporal lobar degeneration. J Neuropathol Exp Neurol. 2016;75(2):175–82.CrossRefPubMed
52.
Zurück zum Zitat Cicognola C, Chiasserini D, Eusebi P, Andreasson U, Vanderstichele H, Zetterberg H, Parnetti L, Blennow K. No diurnal variation of classical and candidate biomarkers of Alzheimer's disease in CSF. Mol Neurodegener. 2016;11(1):65–73.CrossRefPubMedPubMedCentral Cicognola C, Chiasserini D, Eusebi P, Andreasson U, Vanderstichele H, Zetterberg H, Parnetti L, Blennow K. No diurnal variation of classical and candidate biomarkers of Alzheimer's disease in CSF. Mol Neurodegener. 2016;11(1):65–73.CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat Muszynski P, Kulczynska-Przybik A, Borawska R, Litman-Zawadzka A, Slowik A, Klimkowicz-Mrowiec A, Pera J, Dziedzic T, Mroczko B. The relationship between markers of inflammation and degeneration in the central nervous system and the blood-brain barrier impairment in Alzheimer's disease. J Alzheimers Dis. 2017;59(3):903–12.CrossRefPubMed Muszynski P, Kulczynska-Przybik A, Borawska R, Litman-Zawadzka A, Slowik A, Klimkowicz-Mrowiec A, Pera J, Dziedzic T, Mroczko B. The relationship between markers of inflammation and degeneration in the central nervous system and the blood-brain barrier impairment in Alzheimer's disease. J Alzheimers Dis. 2017;59(3):903–12.CrossRefPubMed
54.
Zurück zum Zitat Agustin-Pavon C, Mielcarek M, Garriga-Canut M, Isalan M. Deimmunization for gene therapy: host matching of synthetic zinc finger constructs enables long-term mutant huntingtin repression in mice. Mol Neurodegener. 2016;11(1):64–79.CrossRefPubMedPubMedCentral Agustin-Pavon C, Mielcarek M, Garriga-Canut M, Isalan M. Deimmunization for gene therapy: host matching of synthetic zinc finger constructs enables long-term mutant huntingtin repression in mice. Mol Neurodegener. 2016;11(1):64–79.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Malaisse WJ. Stimulation of insulin release by non-sulfonylurea hypoglycemic agents: the meglitinide family. Horm Metab Res. 1995;27(6):263–6.CrossRefPubMed Malaisse WJ. Stimulation of insulin release by non-sulfonylurea hypoglycemic agents: the meglitinide family. Horm Metab Res. 1995;27(6):263–6.CrossRefPubMed
Metadaten
Titel
Inhibition of DREAM-ATF6 interaction delays onset of cognition deficit in a mouse model of Huntington’s disease
verfasst von
Alejandro López-Hurtado
Daniel F. Burgos
Paz González
Xose M. Dopazo
Valentina González
Alberto Rábano
Britt Mellström
Jose R. Naranjo
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Molecular Brain / Ausgabe 1/2018
Elektronische ISSN: 1756-6606
DOI
https://doi.org/10.1186/s13041-018-0359-6

Weitere Artikel der Ausgabe 1/2018

Molecular Brain 1/2018 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Nicht Creutzfeldt Jakob, sondern Abführtee-Vergiftung

29.05.2024 Hyponatriämie Nachrichten

Eine ältere Frau trinkt regelmäßig Sennesblättertee gegen ihre Verstopfung. Der scheint plötzlich gut zu wirken. Auf Durchfall und Erbrechen folgt allerdings eine Hyponatriämie. Nach deren Korrektur kommt es plötzlich zu progredienten Kognitions- und Verhaltensstörungen.

Schutz der Synapsen bei Alzheimer

29.05.2024 Morbus Alzheimer Nachrichten

Mit einem Neurotrophin-Rezeptor-Modulator lässt sich möglicherweise eine bestehende Alzheimerdemenz etwas abschwächen: Erste Phase-2-Daten deuten auf einen verbesserten Synapsenschutz.

Sozialer Aufstieg verringert Demenzgefahr

24.05.2024 Demenz Nachrichten

Ein hohes soziales Niveau ist mit die beste Versicherung gegen eine Demenz. Noch geringer ist das Demenzrisiko für Menschen, die sozial aufsteigen: Sie gewinnen fast zwei demenzfreie Lebensjahre. Umgekehrt steigt die Demenzgefahr beim sozialen Abstieg.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.