151
|
Benito E, Urbanke H, Ramachandran B, Barth J, Halder R, Awasthi A, Jain G, Capece V, Burkhardt S, Navarro-Sala M, Nagarajan S, Schütz AL, Johnsen SA, Bonn S, Lührmann R, Dean C, Fischer A. HDAC inhibitor-dependent transcriptome and memory reinstatement in cognitive decline models. J Clin Invest 2015; 125:3572-84. [PMID: 26280576 DOI: 10.1172/jci79942] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 07/08/2015] [Indexed: 12/19/2022] Open
Abstract
Aging and increased amyloid burden are major risk factors for cognitive diseases such as Alzheimer's disease (AD). Effective therapies for these diseases are lacking. Here, we evaluated mouse models of age-associated memory impairment and amyloid deposition to study transcriptome and cell type-specific epigenome plasticity in the brain and peripheral organs. We determined that aging and amyloid pathology are associated with inflammation and impaired synaptic function in the hippocampal CA1 region as the result of epigenetic-dependent alterations in gene expression. In both amyloid and aging models, inflammation was associated with increased gene expression linked to a subset of transcription factors, while plasticity gene deregulation was differentially mediated. Amyloid pathology impaired histone acetylation and decreased expression of plasticity genes, while aging altered H4K12 acetylation-linked differential splicing at the intron-exon junction in neurons, but not nonneuronal cells. Furthermore, oral administration of the clinically approved histone deacetylase inhibitor vorinostat not only restored spatial memory, but also exerted antiinflammatory action and reinstated epigenetic balance and transcriptional homeostasis at the level of gene expression and exon usage. This study provides a systems-level investigation of transcriptome plasticity in the hippocampal CA1 region in aging and AD models and suggests that histone deacetylase inhibitors should be further explored as a cost-effective therapeutic strategy against age-associated cognitive decline.
Collapse
|
152
|
Post-occlusion administration of sodium butyrate attenuates cognitive impairment in a rat model of chronic cerebral hypoperfusion. Pharmacol Biochem Behav 2015; 135:53-9. [DOI: 10.1016/j.pbb.2015.05.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 05/07/2015] [Accepted: 05/11/2015] [Indexed: 11/17/2022]
|
153
|
Frick KM. Molecular mechanisms underlying the memory-enhancing effects of estradiol. Horm Behav 2015; 74:4-18. [PMID: 25960081 PMCID: PMC4573242 DOI: 10.1016/j.yhbeh.2015.05.001] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 04/25/2015] [Accepted: 05/01/2015] [Indexed: 11/18/2022]
Abstract
This article is part of a Special Issue "Estradiol and cognition". Since the publication of the 1998 special issue of Hormones and Behavior on estrogens and cognition, substantial progress has been made towards understanding the molecular mechanisms through which 17β-estradiol (E2) regulates hippocampal plasticity and memory. Recent research has demonstrated that rapid effects of E2 on hippocampal cell signaling, epigenetic processes, and local protein synthesis are necessary for E2 to facilitate the consolidation of object recognition and spatial memories in ovariectomized female rodents. These effects appear to be mediated by non-classical actions of the intracellular estrogen receptors ERα and ERβ, and possibly by membrane-bound ERs such as the G-protein-coupled estrogen receptor (GPER). New findings also suggest a key role of hippocampally-synthesized E2 in regulating hippocampal memory formation. The present review discusses these findings in detail and suggests avenues for future study.
Collapse
Affiliation(s)
- Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, 2441 E. Hartford Ave., Milwaukee, WI 53211, USA.
| |
Collapse
|
154
|
Abstract
Apolipoprotein E4 (ApoE4) is a major genetic risk factor for several neurodegenerative disorders, including Alzheimer's disease (AD). Epigenetic dysregulation, including aberrations in histone acetylation, is also associated with AD. We show here for the first time that ApoE4 increases nuclear translocation of histone deacetylases (HDACs) in human neurons, thereby reducing BDNF expression, whereas ApoE3 increases histone 3 acetylation and upregulates BDNF expression. Amyloid-β (Aβ) oligomers, which have been implicated in AD, caused effects similar to ApoE4. Blocking low-density lipoprotein receptor-related protein 1 (LRP-1) receptor with receptor-associated protein (RAP) or LRP-1 siRNA abolished the ApoE effects. ApoE3 also induced expression of protein kinase C ε (PKCε) and PKCε retained HDACs in the cytosol. PKCε activation and ApoE3 supplementation prevented ApoE4-mediated BDNF downregulation. PKCε activation also reversed Aβ oligomer- and ApoE4-induced nuclear import of HDACs, preventing the loss in BDNF. ApoE4 induced HDAC6-BDNF promoter IV binding, which reduced BDNF exon IV expression. Nuclear HDAC4 and HDAC6 were more abundant in the hippocampus of ApoE4 transgenic mice than in ApoE3 transgenic mice or wild-type controls. Nuclear translocation of HDA6 was also elevated in the hippocampus of AD patients compared with age-matched controls. These results provide new insight into the cause of synaptic loss that is the most important pathologic correlate of cognitive deficits in AD.
Collapse
|
155
|
Histone Deacetylase Inhibitor Trichostatin A Ameliorated Endotoxin-Induced Neuroinflammation and Cognitive Dysfunction. Mediators Inflamm 2015; 2015:163140. [PMID: 26273133 PMCID: PMC4530275 DOI: 10.1155/2015/163140] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 03/25/2015] [Accepted: 03/25/2015] [Indexed: 11/19/2022] Open
Abstract
Excessive production of cytokines by microglia may cause cognitive dysfunction and long-lasting behavioral changes. Activating the peripheral innate immune system stimulates cytokine secretion in the central nervous system, which modulates cognitive function. Histone deacetylases (HDACs) modulate cytokine synthesis and release. Trichostatin A (TSA), an HDAC inhibitor, is documented to be anti-inflammatory and neuroprotective. We investigated whether TSA reduces lipopolysaccharide- (LPS-) induced neuroinflammation and cognitive dysfunction. ICR mice were first intraperitoneally (i.p.) injected with vehicle or TSA (0.3 mg/kg). One hour later, they were injected (i.p.) with saline or Escherichia coli LPS (1 mg/kg). We analyzed the food and water intake, body weight loss, and sucrose preference of the injected mice and then determined the microglia activation and inflammatory cytokine expression in the brains of LPS-treated mice and LPS-treated BV-2 microglial cells. In the TSA-pretreated mice, microglial activation was lower, anhedonia did not occur, and LPS-induced cognitive dysfunction (anorexia, weight loss, and social withdrawal) was attenuated. Moreover, mRNA expression of HDAC2, HDAC5, indoleamine 2,3-dioxygenase (IDO), TNF-α, MCP-1, and IL-1β in the brain of LPS-challenged mice and in the LPS-treated BV-2 microglial cells was lower. TSA diminished LPS-induced inflammatory responses in the mouse brain and modulated the cytokine-associated changes in cognitive function, which might be specifically related to reducing HDAC2 and HDAC5 expression.
Collapse
|
156
|
Cuadrado-Tejedor M, Oyarzabal J, Lucas MP, Franco R, García-Osta A. Epigenetic drugs in Alzheimer's disease. Biomol Concepts 2015; 4:433-45. [PMID: 25436752 DOI: 10.1515/bmc-2013-0012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 06/24/2013] [Indexed: 11/15/2022] Open
Abstract
Epigenetic processes, such as DNA methylation and histone acetylation, regulate the genome-environment interactions that may play important roles in a wide range of brain disorders, including Alzheimer's disease (AD). Indeed, the role of epigenetic machinery in learning and memory processes is well documented. In this review, we will focus on the most recent literature on tools that target epigenetic mechanisms, particularly on histone acetylation, and we will discuss the use of chemical probes to validate these targets in therapeutic strategies for AD.
Collapse
|
157
|
Abstract
SIGNIFICANCE Epigenetic inactivation of pivotal genes involved in cell growth is a hallmark of human pathologies, in particular cancer. Histone acetylation balance obtained through opposing actions of histone deacetylases (HDACs) and histone acetyltransferases is one epigenetic mechanism controlling gene expression and is, thus, associated with disease etiology and progression. Interfering pharmacologically with HDAC activity can correct abnormalities in cell proliferation, migration, vascularization, and death. RECENT ADVANCES Histone deacetylase inhibitors (HDACi) represent a new class of cytostatic agents that interfere with the function of HDACs and are able to increase gene expression by indirectly inducing histone acetylation. Several HDACi, alone or in combination with DNA-demethylating agents, chemopreventive, or classical chemotherapeutic drugs, are currently being used in clinical trials for solid and hematological malignancies, and are, thus, promising candidates for cancer therapy. CRITICAL ISSUES (i) Non-specific (off-target) HDACi effects due to activities unassociated with HDAC inhibition. (ii) Advantages/disadvantages of non-selective or isoform-directed HDACi. (iii) Limited number of response-predictive biomarkers. (iv) Toxicity leading to dysfunction of critical biological processes. FUTURE DIRECTIONS Selective HDACi could achieve enhanced clinical utility by reducing or eliminating the serious side effects associated with current first-generation non-selective HDACi. Isoform-selective and pan-HDACi candidates might benefit from the identification of biomarkers, enabling better patient stratification and prediction of response to treatment.
Collapse
Affiliation(s)
- Rosaria Benedetti
- 1 Department of Biochemistry, Biophysics, and General Pathology, Seconda Università degli Studi di Napoli , Napoli, Italy
| | - Mariarosaria Conte
- 1 Department of Biochemistry, Biophysics, and General Pathology, Seconda Università degli Studi di Napoli , Napoli, Italy
| | - Lucia Altucci
- 1 Department of Biochemistry, Biophysics, and General Pathology, Seconda Università degli Studi di Napoli , Napoli, Italy .,2 Istituto di Genetica e Biofisica "Adriano Buzzati-Traverso," Napoli, Italy
| |
Collapse
|
158
|
Lu X, Wang L, Yu C, Yu D, Yu G. Histone Acetylation Modifiers in the Pathogenesis of Alzheimer's Disease. Front Cell Neurosci 2015; 9:226. [PMID: 26136662 PMCID: PMC4468862 DOI: 10.3389/fncel.2015.00226] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 05/29/2015] [Indexed: 01/07/2023] Open
Abstract
It is becoming more evident that histone acetylation, as one of the epigenetic modifications or markers, plays a key role in the etiology of Alzheimer’s disease (AD). Histone acetylases and histone deacetylases (HDACs) are the well-known covalent enzymes that modify the reversible acetylation of lysine residues in histone amino-terminal domains. In AD, however, the roles of these enzymes are controversial. Some recent studies indicate that HDAC inhibitors are neuroprotective by regulating memory and synaptic dysfunctions in cellular and animal models of AD; while on the other hand, increase of histone acetylation have been implicated in AD pathology. In this review, we focus on the recent advances on the roles of histone acetylation covalent enzymes in AD and discuss how targeting these enzymes can ultimately lead to therapeutic approaches for treating AD.
Collapse
Affiliation(s)
- Xi Lu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University , Chongqing , China
| | - Li Wang
- Department of Biotherapy and Hemato-oncology, Chongqing Cancer Institute , Chongqing , China
| | - Caijia Yu
- The Commonwealth Medical College , Scranton, PA , USA
| | - Daohai Yu
- Department of Clinical Sciences, Temple Clinical Research Institute, Temple University School of Medicine , Philadelphia, PA , USA
| | - Gang Yu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University , Chongqing , China
| |
Collapse
|
159
|
Abstract
With ever-increasing elder population, the high incidence of age-related diseases such as neurodegenerative disorders has turned out to be a huge public concern. Especially the elders and their families dreadfully suffer from the learning, behavioral and cognitive impairments. The lack of effective therapies for such a horrible symptom makes a great demanding for biological mechanism study for cognitive aging. Epigenetics is an emerging field that broadens the dimensions of mammalian genome blueprint. It is, unlike genetics, not only inheritable but also reversible. Recent studies suggest that DNA methylation, one of major epigenetic mechanisms, plays a pivotal role in the pathogenesis of age-related neurodegenerations and cognitive defects. In this review, the evolving knowledge of age-related cognitive functions and the potential DNA methylation mechanism of cognitive aging are discussed. That indicates the impairment of DNA methylation may be a crucial but reversible mechanism of behavioral and cognitive related neurodegeneration. The methods to examine the dynamics of DNA methylation patterns at tissue and single cell level and at the representative scale as well as the whole genome single base resolution are also briefly discussed. Importantly, the challenges of DNA methylation mechanism of cognitive aging research are brought up, and the possible solutions to tackle these difficulties are put forward.
Collapse
Affiliation(s)
- Xiangru Xu
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
160
|
HDAC inhibitors as cognitive enhancers in fear, anxiety and trauma therapy: where do we stand? Biochem Soc Trans 2015; 42:569-81. [PMID: 24646280 PMCID: PMC3961057 DOI: 10.1042/bst20130233] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A novel strategy to treat anxiety and fear-related disorders such as phobias, panic and PTSD (post-traumatic stress disorder) is combining CBT (cognitive behavioural therapy), including extinction-based exposure therapy, with cognitive enhancers. By targeting and boosting mechanisms underlying learning, drug development in this field aims at designing CBT-augmenting compounds that help to overcome extinction learning deficits, promote long-term fear inhibition and thus support relapse prevention. Progress in revealing the role of epigenetic regulation of specific genes associated with extinction memory generation has opened new avenues in this direction. The present review examines recent evidence from pre-clinical studies showing that increasing histone acetylation, either via genetic or pharmacological inhibition of HDACs (histone deacetylases) by e.g. vorinostat/SAHA (suberoylanilide hydroxamic acid), entinostat/MS-275, sodium butyrate, TSA (trichostatin A) or VPA (valproic acid), or by targeting HATs (histone acetyltransferases), augments fear extinction and, importantly, generates a long-term extinction memory that can protect from return of fear phenomena. The molecular mechanisms and pathways involved including BDNF (brain-derived neurotrophic factor) and NMDA (N-methyl-D-aspartate) receptor signalling are just beginning to be revealed. First studies in healthy humans are in support of extinction-facilitating effects of HDAC inhibitors. Very recent evidence that HDAC inhibitors can rescue deficits in extinction-memory-impaired rodents indicates a potential clinical utility of this approach also for exposure therapy-resistant patients. Important future work includes investigation of the long-term safety aspects of HDAC inhibitor treatment, as well as design of isotype(s)-specific inhibitors. Taken together, HDAC inhibitors display promising potential as pharmacological adjuncts to augment the efficacy of exposure-based approaches in anxiety and trauma therapy.
Collapse
|
161
|
Kraepelin revisited: schizophrenia from degeneration to failed regeneration. Mol Psychiatry 2015; 20:671-6. [PMID: 25824303 DOI: 10.1038/mp.2015.35] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 02/11/2015] [Accepted: 02/24/2015] [Indexed: 12/18/2022]
Abstract
One hundred years after its conceptual definition as 'Dementia Praecox' by Emil Kraepelin, schizophrenia is still a serious psychiatric illness that affects young adults and leads to disability in at least half of patients. The key treatment issue is partial or non-response, especially of negative symptoms. The illness is also associated with different degrees of cognitive dysfunction, particularly in verbal and working memory; the resulting functional impairment may lead to unemployment and an inability to maintain stable relationships. Patients' cognitive dysfunction led Kraepelin to the assumption that schizophrenia is a form of juvenile dementia caused by a degenerative process of the human brain. Postmortem studies and a plethora of imaging studies do not support the notion of a degenerative process, but such a process is supported by the recently published, largest genome-wide association study on schizophrenia. More than a 100 hits were described, converging on pathways that have a significant role in dopamine metabolism in immune modulation, calcium signalling and synaptic plasticity. This review suggests that research should focus on animal models based on risk genes like transcription factor 4 and study the effects of exposure to environmental stressors relevant for schizophrenia. The use of relevant end points like pre-pulse inhibition or cognitive dysfunction will allow us to gain an understanding of the molecular pathways in schizophrenia and consequently result in improved treatment options, especially for the disabling aspects of this illness.
Collapse
|
162
|
Szyf M. Prospects for the development of epigenetic drugs for CNS conditions. Nat Rev Drug Discov 2015; 14:461-74. [DOI: 10.1038/nrd4580] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
163
|
Szyf M. Epigenetics, a key for unlocking complex CNS disorders? Therapeutic implications. Eur Neuropsychopharmacol 2015; 25:682-702. [PMID: 24857313 DOI: 10.1016/j.euroneuro.2014.01.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 12/07/2013] [Accepted: 01/11/2014] [Indexed: 12/13/2022]
Abstract
Aberrant changes in gene function are believed to be involved in a wide spectrum of human disease including behavioral, cognitive and neurodegenerative pathologies. Most of the attention in last few decades have focused on changes in gene sequence as a cause of gene dysfunction leading to disease and mental health disorders. Germ line mutations or other alterations in the sequence of DNA that associate with different behavioral and neurological pathologies have been identified. However, sequence alterations explain only a small fraction of the cases. In addition there is evidence for "gene-environment" interactions in the brain suggesting mechanisms that alter gene function and the phenotype through environmental exposure. Genes are programmed by "epigenetic" mechanisms such as chromatin structure, chromatin modification and DNA methylation. These mechanisms confer on similar sequences different identities during cellular differentiation. Epigenetic differences are proposed to be involved in differentiating gene function in response to different environmental contexts and could result in alterations in functional gene networks that lead to brain disease. Epigenetic markers could serve important biomarkers in brain and behavioral diseases. Moreover, epigenetic processes are potentially reversible pointing to epigenetic therapeutics in psychotherapy.
Collapse
Affiliation(s)
- Moshe Szyf
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada H3G1Y5.
| |
Collapse
|
164
|
Valor LM. Epigenetic-based therapies in the preclinical and clinical treatment of Huntington's disease. Int J Biochem Cell Biol 2015; 67:45-8. [PMID: 25936670 DOI: 10.1016/j.biocel.2015.04.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/17/2015] [Accepted: 04/21/2015] [Indexed: 01/08/2023]
Abstract
The study of epigenetics is providing novel insights about the functional and developmental complexity of the nervous system. In neuropathology, therapies aimed at correcting epigenetic dysregulation have been extensively documented in a large variety of models for neurodegenerative, neurodevelopmental and psychiatric disorders. Taking the treatment of Huntington's disease as a paradigm for the study of these ameliorative strategies, this review updates the main conclusions derived from the use of epigenetic drugs at the preclinical and clinical stages, including actions beyond epigenetics. This article is part of a Directed Issue entitled: Epigenetics dynamics in development and disease.
Collapse
Affiliation(s)
- Luis M Valor
- Instituto de Neurociencias de Alicante (Universidad Miguel Hernández - Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant, 03550 Alicante, Spain.
| |
Collapse
|
165
|
Bang SR, Ambavade SD, Jagdale PG, Adkar PP, Waghmare AB, Ambavade PD. Lacosamide reduces HDAC levels in the brain and improves memory: Potential for treatment of Alzheimer's disease. Pharmacol Biochem Behav 2015; 134:65-9. [PMID: 25931268 DOI: 10.1016/j.pbb.2015.04.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 04/10/2015] [Accepted: 04/15/2015] [Indexed: 11/28/2022]
Abstract
Lacosamide, a histone deacetylase (HDAC) inhibitor, has been approved for the treatment of epilepsy. Some HDAC inhibitors have been proven effective for the treatment of memory disorders. The present investigation was designed to evaluate the effect of lacosamide on memory and brain HDAC levels. The effect on memory was evaluated in animals with scopolamine-induced amnesia using the elevated plus maze, object recognition test, and radial arm maze. The levels of acetylcholinesterase and HDAC in the cerebral cortex were evaluated. Lacosamide at doses of 10 and 30mg/kg significantly reduced the transfer latency in the elevated plus maze. Lacosamide at a dose of 30mg/kg significantly increased the time spent with a familiar object in the object recognition test at the 24h interval and decreased the time spent in the baited arm. Moreover, at this dose, the number of errors in the radial arm maze at 3 and 24h intervals was minimized and a reduction in the level of HDAC1, but not acetylcholinesterase, was observed in the cerebral cortex. These effects of lacosamide are equivalent to those of piracetam at a dose of 300mg/kg. These results suggest that lacosamide at a 30mg/kg dose improves disrupted memory, possibly by inhibiting HDAC, and could be used to treat amnesic symptoms of Alzheimer's disease.
Collapse
Affiliation(s)
- Shraddha R Bang
- Department of Pharmacology, JSPM's Jayawantrao Sawant College of Pharmacy and Research(1), Handewadi Road, Hadapsar, Pune 411028, India
| | - Shirishkumar D Ambavade
- Department of Pharmacology, JSPM's Jayawantrao Sawant College of Pharmacy and Research(1), Handewadi Road, Hadapsar, Pune 411028, India.
| | - Priti G Jagdale
- Department of Pharmacology, JSPM's Jayawantrao Sawant College of Pharmacy and Research(1), Handewadi Road, Hadapsar, Pune 411028, India
| | - Prafulla P Adkar
- Department of Pharmacology, JSPM's Jayawantrao Sawant College of Pharmacy and Research(1), Handewadi Road, Hadapsar, Pune 411028, India
| | - Arun B Waghmare
- Haffkine Biopharmaceutical Corporation Ltd., Pimpri, Pune 18, India
| | | |
Collapse
|
166
|
Effects of sodium butyrate on aversive memory in rats submitted to sepsis. Neurosci Lett 2015; 595:134-8. [PMID: 25888815 DOI: 10.1016/j.neulet.2015.04.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 03/31/2015] [Accepted: 04/10/2015] [Indexed: 01/08/2023]
Abstract
Epigenetic mechanisms are involved in normal behavior and are implicated in several brain neurodegenerative conditions, psychiatric and inflammatory diseases as well. Moreover, it has been demonstrated that sepsis lead to an imbalance in acetylation of histones and that histone deacetylase inhibitors (HDACi) can reverse this condition. In the present study, we evaluated the effects of a microinjection of sodium butyrate (SB, HDACi) into cerebral ventricle on aversive memory in rats submitted to the sepsis. Rats were given a single intraventricular injection of artificial cerebrospinal fluid (ACSF) or SB and immediately after the stereotaxic surgery and the drug infusion, the animals were subjected to cecal ligation and perforation (CLP). The animals were killed twenty four hours or ten days after sepsis induction and the prefrontal cortex, hippocampus, striatum and cortex were obtained to the determination of histone deacetylase activity. In a separate cohort of animals 10 days after sepsis induction, it was performed the inhibitory avoidance task. SB administration was able to reverse the impairment in aversive memory and inhibited the HDAC activity in prefrontal cortex and hippocampus 10 days after CLP. These support a role for an epigenetic mechanism in the long-term cognitive impairments observed in sepsis survivors animals.
Collapse
|
167
|
(-)-Epigallocatechin-3-gallate attenuates cognitive deterioration in Alzheimer's disease model mice by upregulating neprilysin expression. Exp Cell Res 2015; 334:136-45. [PMID: 25882496 DOI: 10.1016/j.yexcr.2015.04.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 03/27/2015] [Accepted: 04/05/2015] [Indexed: 01/04/2023]
Abstract
Epigenetic changes are involved in learning and memory, and histone deacetylase (HDAC) inhibitors are considered potential therapeutic agents for Alzheimer's disease (AD). We previously reported that (-)-epigallocatechin-3-gallate (EGCG) acts as an HDAC inhibitor. Here, we demonstrate that EGCG reduced β-amyloid (Aβ) accumulation in vitro and rescued cognitive deterioration in senescence-accelerated mice P8 (SAMP8) via intragastric administration of low- and high-dose EGCG (5 and 15 mg/kg, respectively) for 60 days. The AD brain has decreased levels of the rate-limiting degradation enzyme of Aβ, neprilysin (NEP). We found an association between EGCG-induced reduction in Aβ accumulation and elevated NEP expression. Further, NEP silencing prevented the EGCG-induced Aβ downregulation. Our findings suggest that EGCG might be effective for treating AD.
Collapse
|
168
|
Morishita H, Kundakovic M, Bicks L, Mitchell A, Akbarian S. Interneuron epigenomes during the critical period of cortical plasticity: Implications for schizophrenia. Neurobiol Learn Mem 2015; 124:104-10. [PMID: 25849095 DOI: 10.1016/j.nlm.2015.03.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 03/23/2015] [Accepted: 03/25/2015] [Indexed: 10/23/2022]
Abstract
Schizophrenia, a major psychiatric disorder defined by delusions and hallucinations, among other symptoms, often with onset in early adulthood, is potentially associated with molecular and cellular alterations in parvalbumin-expressing fast spiking interneurons and other constituents of the cortical inhibitory GABAergic circuitry. The underlying mechanisms, including the role of disease-associated risk factors operating in adolescence such as drug abuse and social stressors, remain incompletely understood. Here, we summarize emerging findings from animal models, highlighting the ability of parvalbuminergic interneurons (PVI) to induce, during the juvenile period, long-term plastic changes in prefrontal and visual cortex, thereby altering perception, cognition and behavior in the adult. Of note, molecular alterations in PVI from subjects with schizophrenia, including downregulated expression of a subset of GABAergic genes, have also been found in juvenile stress models of the disorder. Some of the transcriptional alterations observed in schizophrenia postmortem brain could be linked to changes in the epigenetic architecture of GABAergic gene promoters, including dysregulated DNA methylation, histone modification patterns and disruption of promoter-enhancer interactions at site of chromosomal loop formations. Therefore, we predict that, in the not-to-distant future, PVI- and other cell-type specific epigenomic mappings in the animal model and human brain will provide novel insights into the pathophysiology of schizophrenia and related psychotic diseases, including the role of cortical GABAergic circuitry in shaping long-term plasticity and cognitive function of the cerebral cortex.
Collapse
Affiliation(s)
- Hirofumi Morishita
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States; Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, United States; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, United States.
| | - Marija Kundakovic
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, United States
| | - Lucy Bicks
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, United States
| | - Amanda Mitchell
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, United States
| | - Schahram Akbarian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, United States.
| |
Collapse
|
169
|
Fleiss B, Tann CJ, Degos V, Sigaut S, Van Steenwinckel J, Schang AL, Kichev A, Robertson NJ, Mallard C, Hagberg H, Gressens P. Inflammation-induced sensitization of the brain in term infants. Dev Med Child Neurol 2015; 57 Suppl 3:17-28. [PMID: 25800488 DOI: 10.1111/dmcn.12723] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/24/2014] [Indexed: 12/12/2022]
Abstract
Perinatal insults are a leading cause of infant mortality and amongst survivors are frequently associated with neurocognitive impairment, cerebral palsy (CP), and seizure disorders. The events leading to perinatal brain injury are multifactorial. This review describes how one subinjurious factor affecting the brain sensitizes it to a second injurious factor, causing an exacerbated injurious cascade. We will review the clinical and experimental evidence, including observations of high rates of maternal and fetal infections in term-born infants with neonatal encephalopathy and cerebral palsy. In addition, we will discuss preclinical evidence for the sensitizing effects of inflammation on injuries, such as hypoxia-ischaemia, our current understanding of the mechanisms underpinning the sensitization process, and the possibility for neuroprotection.
Collapse
Affiliation(s)
- Bobbi Fleiss
- Inserm, U1141, Paris, France; University Paris Diderot, Sorbonne Paris Cité, UMRS 1141, Paris, France; Department of Child Neurology, APHP, Robert Debré Hospital, Paris, France; PremUP, Paris, France; Division of Imaging Sciences, Department of Perinatal Imaging and Health, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
170
|
Post-training, intrahippocampal HDAC inhibition differentially impacts neural circuits underlying spatial memory in adult and aged mice. Hippocampus 2015; 25:827-37. [DOI: 10.1002/hipo.22406] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2014] [Indexed: 11/07/2022]
|
171
|
Fukui T, Asakura K, Hikichi C, Ishikawa T, Murai R, Hirota S, Murate KI, Kizawa M, Ueda A, Ito S, Mutoh T. Histone deacetylase inhibitor attenuates neurotoxicity of clioquinol in PC12 cells. Toxicology 2015; 331:112-8. [PMID: 25758465 DOI: 10.1016/j.tox.2015.01.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 01/19/2015] [Accepted: 01/20/2015] [Indexed: 12/25/2022]
Abstract
Clioquinol is considered to be a causative agent of subacute myelo-optico neuropathy (SMON), although the pathogenesis of SMON is yet to be elucidated. We have previously shown that clioquinol inhibits nerve growth factor (NGF)-induced Trk autophosphorylation in PC12 cells transformed with human Trk cDNA. To explore the further mechanism of neuronal damage by clioquinol, we evaluated the acetylation status of histones in PC12 cells. Clioquinol reduced the level of histone acetylation, and the histone deacetylase (HDAC) inhibitor Trichostatin A upregulated acetylated histones and prevented the neuronal cell damage caused by clioquinol. In addition, treatment with HDAC inhibitor decreased neurite retraction and restored the inhibition of NGF-induced Trk autophosphorylation by clioquinol. Thus, clioquinol induced neuronal cell death via deacetylation of histones, and HDAC inhibitor alleviates the neurotoxicity of clioquinol. Clioquinol is now used as a potential medicine for malignancies and neurodegenerative diseases. Therefore, HDAC inhibitors can be used as a candidate medicine for the prevention of its side effects on neuronal cells.
Collapse
Affiliation(s)
- Takao Fukui
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Kunihiko Asakura
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi 470-1192, Japan.
| | - Chika Hikichi
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Tomomasa Ishikawa
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Rie Murai
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Seiko Hirota
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Ken-Ichiro Murate
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Madoko Kizawa
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Akihiro Ueda
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Shinji Ito
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Tatsuro Mutoh
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi 470-1192, Japan
| |
Collapse
|
172
|
Mielcarek M, Zielonka D, Carnemolla A, Marcinkowski JT, Guidez F. HDAC4 as a potential therapeutic target in neurodegenerative diseases: a summary of recent achievements. Front Cell Neurosci 2015; 9:42. [PMID: 25759639 PMCID: PMC4338808 DOI: 10.3389/fncel.2015.00042] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 01/28/2015] [Indexed: 12/13/2022] Open
Abstract
For the past decade protein acetylation has been shown to be a crucial post-transcriptional modification involved in the regulation of protein functions. Histone acetyltransferases (HATs) mediate acetylation of histones which results in the nucleosomal relaxation associated with gene expression. The reverse reaction, histone deacetylation, is mediated by histone deacetylases (HDACs) leading to chromatin condensation followed by transcriptional repression. HDACs are divided into distinct classes: I, IIa, IIb, III, and IV, on the basis of size and sequence homology, as well as formation of distinct repressor complexes. Implications of HDACs in many diseases, such as cancer, heart failure, and neurodegeneration, have identified these molecules as unique and attractive therapeutic targets. The emergence of HDAC4 among the members of class IIa family as a major player in synaptic plasticity raises important questions about its functions in the brain. The characterization of HDAC4 specific substrates and molecular partners in the brain will not only provide a better understanding of HDAC4 biological functions but also might help to develop new therapeutic strategies to target numerous malignancies. In this review we highlight and summarize recent achievements in understanding the biological role of HDAC4 in neurodegenerative processes.
Collapse
Affiliation(s)
- Michal Mielcarek
- Department of Medical and Molecular Genetics, King's College London London, UK
| | - Daniel Zielonka
- Department of Social Medicine, Poznan University of Medical Sciences Poznan, Poland
| | - Alisia Carnemolla
- Department of Medical and Molecular Genetics, King's College London London, UK
| | - Jerzy T Marcinkowski
- Department of Social Medicine, Poznan University of Medical Sciences Poznan, Poland
| | - Fabien Guidez
- INSERM UMRS 1131, Université Paris Diderot, Institut Universitaire d'hématologie (IUH), Hôpital Saint-Louis Paris, France
| |
Collapse
|
173
|
Singewald N, Schmuckermair C, Whittle N, Holmes A, Ressler KJ. Pharmacology of cognitive enhancers for exposure-based therapy of fear, anxiety and trauma-related disorders. Pharmacol Ther 2014; 149:150-90. [PMID: 25550231 PMCID: PMC4380664 DOI: 10.1016/j.pharmthera.2014.12.004] [Citation(s) in RCA: 275] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 12/24/2014] [Indexed: 12/20/2022]
Abstract
Pathological fear and anxiety are highly debilitating and, despite considerable advances in psychotherapy and pharmacotherapy they remain insufficiently treated in many patients with PTSD, phobias, panic and other anxiety disorders. Increasing preclinical and clinical evidence indicates that pharmacological treatments including cognitive enhancers, when given as adjuncts to psychotherapeutic approaches [cognitive behavioral therapy including extinction-based exposure therapy] enhance treatment efficacy, while using anxiolytics such as benzodiazepines as adjuncts can undermine long-term treatment success. The purpose of this review is to outline the literature showing how pharmacological interventions targeting neurotransmitter systems including serotonin, dopamine, noradrenaline, histamine, glutamate, GABA, cannabinoids, neuropeptides (oxytocin, neuropeptides Y and S, opioids) and other targets (neurotrophins BDNF and FGF2, glucocorticoids, L-type-calcium channels, epigenetic modifications) as well as their downstream signaling pathways, can augment fear extinction and strengthen extinction memory persistently in preclinical models. Particularly promising approaches are discussed in regard to their effects on specific aspects of fear extinction namely, acquisition, consolidation and retrieval, including long-term protection from return of fear (relapse) phenomena like spontaneous recovery, reinstatement and renewal of fear. We also highlight the promising translational value of the preclinial research and the clinical potential of targeting certain neurochemical systems with, for example d-cycloserine, yohimbine, cortisol, and L-DOPA. The current body of research reveals important new insights into the neurobiology and neurochemistry of fear extinction and holds significant promise for pharmacologically-augmented psychotherapy as an improved approach to treat trauma and anxiety-related disorders in a more efficient and persistent way promoting enhanced symptom remission and recovery.
Collapse
Affiliation(s)
- N Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, Leopold-Franzens University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria.
| | - C Schmuckermair
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, Leopold-Franzens University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - N Whittle
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, Leopold-Franzens University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - A Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, USA
| | - K J Ressler
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
174
|
Takuma K, Hara Y, Kataoka S, Kawanai T, Maeda Y, Watanabe R, Takano E, Hayata-Takano A, Hashimoto H, Ago Y, Matsuda T. Chronic treatment with valproic acid or sodium butyrate attenuates novel object recognition deficits and hippocampal dendritic spine loss in a mouse model of autism. Pharmacol Biochem Behav 2014; 126:43-9. [DOI: 10.1016/j.pbb.2014.08.013] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 08/21/2014] [Accepted: 08/30/2014] [Indexed: 11/24/2022]
|
175
|
Stilling RM, Bordenstein SR, Dinan TG, Cryan JF. Friends with social benefits: host-microbe interactions as a driver of brain evolution and development? Front Cell Infect Microbiol 2014; 4:147. [PMID: 25401092 PMCID: PMC4212686 DOI: 10.3389/fcimb.2014.00147] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 10/03/2014] [Indexed: 12/21/2022] Open
Abstract
The tight association of the human body with trillions of colonizing microbes that we observe today is the result of a long evolutionary history. Only very recently have we started to understand how this symbiosis also affects brain function and behavior. In this hypothesis and theory article, we propose how host-microbe associations potentially influenced mammalian brain evolution and development. In particular, we explore the integration of human brain development with evolution, symbiosis, and RNA biology, which together represent a “social triangle” that drives human social behavior and cognition. We argue that, in order to understand how inter-kingdom communication can affect brain adaptation and plasticity, it is inevitable to consider epigenetic mechanisms as important mediators of genome-microbiome interactions on an individual as well as a transgenerational time scale. Finally, we unite these interpretations with the hologenome theory of evolution. Taken together, we propose a tighter integration of neuroscience fields with host-associated microbiology by taking an evolutionary perspective.
Collapse
Affiliation(s)
- Roman M Stilling
- Alimentary Pharmabiotic Centre, University College Cork Cork, Ireland ; Department Anatomy and Neuroscience, University College Cork Cork, Ireland
| | - Seth R Bordenstein
- Departments of Biological Sciences and Pathology, Microbiology, and Immunology, Vanderbilt University Nashville, TN, USA
| | - Timothy G Dinan
- Alimentary Pharmabiotic Centre, University College Cork Cork, Ireland ; Department of Psychiatry, University College Cork Cork, Ireland
| | - John F Cryan
- Alimentary Pharmabiotic Centre, University College Cork Cork, Ireland ; Department Anatomy and Neuroscience, University College Cork Cork, Ireland
| |
Collapse
|
176
|
Schroeder FA, Wang C, Van de Bittner GC, Neelamegam R, Takakura WR, Karunakaran A, Wey HY, Reis SA, Gale J, Zhang YL, Holson EB, Haggarty SJ, Hooker JM. PET imaging demonstrates histone deacetylase target engagement and clarifies brain penetrance of known and novel small molecule inhibitors in rat. ACS Chem Neurosci 2014; 5:1055-62. [PMID: 25188794 PMCID: PMC4198064 DOI: 10.1021/cn500162j] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
![]()
Histone deacetylase (HDAC) enzymes
have been demonstrated as critical
components in maintaining chromatin homeostasis, CNS development,
and normal brain function. Evidence in mouse models links HDAC expression
to learning, memory, and mood-related behaviors; small molecule HDAC
inhibitor tool compounds have been used to demonstrate the importance
of specific HDAC subtypes in modulating CNS-disease-related behaviors
in rodents. So far, no direct evidence exists to understand the quantitative
changes in HDAC target engagement that are necessary to alter biochemistry
and behavior in a living animal. Understanding the relationship between
target engagement and in vivo effect is essential
in refining new ways to alleviate disease. We describe here, using
positron emission tomography (PET) imaging of rat brain, the in vivo target engagement of a subset of class I/IIb HDAC
enzymes implicated in CNS-disease (HDAC subtypes 1, 2, 3, and 6).
We found marked differences in the brain penetrance of tool compounds
from the hydroxamate and benzamide HDAC inhibitor classes and resolved
a novel, highly brain penetrant benzamide, CN147, chronic treatment
with which resulted in an antidepressant-like effect in a rat behavioral
test. Our work highlights a new translational path for understanding
the molecular and behavioral consequences of HDAC target engagement.
Collapse
Affiliation(s)
- F. A. Schroeder
- Athinoula
A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
- Chemical
Neurobiology Laboratory, Departments of Neurology and Psychiatry,
Center for Human Genetic Research, Massachusetts General Hospital, 185
Cambridge Street, Boston, Massachusetts 02114, United States
| | - C. Wang
- Athinoula
A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - G. C. Van de Bittner
- Athinoula
A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - R. Neelamegam
- Athinoula
A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - W. R. Takakura
- Athinoula
A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - A. Karunakaran
- Athinoula
A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - H. Y. Wey
- Athinoula
A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - S. A. Reis
- Chemical
Neurobiology Laboratory, Departments of Neurology and Psychiatry,
Center for Human Genetic Research, Massachusetts General Hospital, 185
Cambridge Street, Boston, Massachusetts 02114, United States
| | - J. Gale
- Stanley
Center for Psychiatric Research, Broad Institute of Harvard and MIT, 7
Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - Y. L. Zhang
- Stanley
Center for Psychiatric Research, Broad Institute of Harvard and MIT, 7
Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - E. B. Holson
- Stanley
Center for Psychiatric Research, Broad Institute of Harvard and MIT, 7
Cambridge Center, Cambridge, Massachusetts 02142, United States
| | - S. J. Haggarty
- Chemical
Neurobiology Laboratory, Departments of Neurology and Psychiatry,
Center for Human Genetic Research, Massachusetts General Hospital, 185
Cambridge Street, Boston, Massachusetts 02114, United States
| | - J. M. Hooker
- Athinoula
A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| |
Collapse
|
177
|
Aurones as histone deacetylase inhibitors: identification of key features. Bioorg Med Chem Lett 2014; 24:5497-501. [PMID: 25455492 DOI: 10.1016/j.bmcl.2014.10.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 09/29/2014] [Accepted: 10/01/2014] [Indexed: 02/02/2023]
Abstract
In this study, a total of 22 flavonoids were tested for their HDAC inhibitory activity using fluorimetric and BRET-based assays. Four aurones were found to be active in both assays and showed IC50 values below 20 μM in the enzymatic assay. Molecular modelling revealed that the presence of hydroxyl groups was responsible for good compound orientation within the isoenzyme catalytic site and zinc chelation.
Collapse
|
178
|
Fortress AM, Frick KM. Epigenetic regulation of estrogen-dependent memory. Front Neuroendocrinol 2014; 35:530-49. [PMID: 24878494 PMCID: PMC4174980 DOI: 10.1016/j.yfrne.2014.05.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 05/15/2014] [Accepted: 05/16/2014] [Indexed: 02/09/2023]
Abstract
Hippocampal memory formation is highly regulated by post-translational histone modifications and DNA methylation. Accordingly, these epigenetic processes play a major role in the effects of modulatory factors, such as sex steroid hormones, on hippocampal memory. Our laboratory recently demonstrated that the ability of the potent estrogen 17β-estradiol (E2) to enhance hippocampal-dependent novel object recognition memory in ovariectomized female mice requires ERK-dependent histone H3 acetylation and DNA methylation in the dorsal hippocampus. Although these data provide valuable insight into the chromatin modifications that mediate the memory-enhancing effects of E2, epigenetic regulation of gene expression is enormously complex. Therefore, more research is needed to fully understand how E2 and other hormones employ epigenetic alterations to shape behavior. This review discusses the epigenetic alterations shown thus far to regulate hippocampal memory, briefly reviews the effects of E2 on hippocampal function, and describes in detail our work on epigenetic regulation of estrogenic memory enhancement.
Collapse
Affiliation(s)
- Ashley M Fortress
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States.
| |
Collapse
|
179
|
Lopez-Atalaya JP, Barco A. Can changes in histone acetylation contribute to memory formation? Trends Genet 2014; 30:529-39. [PMID: 25269450 DOI: 10.1016/j.tig.2014.09.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 09/04/2014] [Accepted: 09/05/2014] [Indexed: 01/31/2023]
Abstract
Neuronal histone acetylation has been postulated to be a mnemonic substrate and a target for memory enhancers and neuropsychiatric drugs. Here we critically evaluate this view and examine the apparent conflict between the proposed instructive role for histone acetylation in memory-related transcription and the insights derived from genomic and genetic studies in other systems. We next discuss the suitability of activity-dependent neuronal histone acetylation as a mnemonic substrate and debate alternative interpretations of current evidence. We believe that further progress in our understanding of the role of histone acetylation and other epigenetic modifications in neuronal plasticity, memory, and neuropsychiatric disorders requires a clear discrimination between cause and effect so that novel epigenetics-related processes can be distinguished from classical transcriptional mechanisms.
Collapse
Affiliation(s)
- Jose P Lopez-Atalaya
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas, Av. Santiago Ramón y Cajal s/n. Sant Joan d'Alacant, 03550 Alicante, Spain
| | - Angel Barco
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas, Av. Santiago Ramón y Cajal s/n. Sant Joan d'Alacant, 03550 Alicante, Spain.
| |
Collapse
|
180
|
Mahgoub M, Monteggia LM. A role for histone deacetylases in the cellular and behavioral mechanisms underlying learning and memory. ACTA ACUST UNITED AC 2014; 21:564-8. [PMID: 25227251 PMCID: PMC4175496 DOI: 10.1101/lm.036012.114] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Histone deacetylases (HDACs) are a family of chromatin remodeling enzymes that restrict access of transcription factors to the DNA, thereby repressing gene expression. In contrast, histone acetyltransferases (HATs) relax the chromatin structure allowing for an active chromatin state and promoting gene transcription. Accumulating data have demonstrated a crucial function for histone acetylation and histone deacetylation in regulating the cellular and behavioral mechanisms underlying synaptic plasticity and learning and memory. In trying to delineate the roles of individual HDACs, genetic tools have been used to manipulate HDAC expression in rodents, uncovering distinct contributions of individual HDACs in regulating the processes of memory formation. Moreover, recent findings have suggested an important role for HDAC inhibitors in enhancing learning and memory processes as well as ameliorating symptoms related to neurodegenerative diseases. In this review, we focus on the role of HDACs in learning and memory, as well as significant data emerging from the field in support of HDAC inhibitors as potential therapeutic targets for the treatment of cognitive disorders.
Collapse
Affiliation(s)
- Melissa Mahgoub
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75235-9111, USA
| | - Lisa M Monteggia
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75235-9111, USA
| |
Collapse
|
181
|
Brunkhorst R, Vutukuri R, Pfeilschifter W. Fingolimod for the treatment of neurological diseases-state of play and future perspectives. Front Cell Neurosci 2014; 8:283. [PMID: 25309325 PMCID: PMC4162362 DOI: 10.3389/fncel.2014.00283] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 08/25/2014] [Indexed: 11/25/2022] Open
Abstract
Sphingolipids are a fascinating class of signaling molecules derived from the membrane lipid sphingomyelin. They show abundant expression in the brain. Complex sphingolipids such as glycosphingolipids (gangliosides and cerebrosides) regulate vesicular transport and lysosomal degradation and their dysregulation can lead to storage diseases with a neurological phenotype. More recently, simple sphingolipids such ceramide, sphingosine and sphingosine 1-phosphate (S1P) were discovered to signal in response to many extracellular stimuli. Forming an intricate signaling network, the balance of these readily interchangeable mediators is decisive for cell fate under stressful conditions. The immunomodulator fingolimod is the prodrug of an S1P receptor agonist. Following receptor activation, the drug leads to downregulation of the S1P1 receptor inducing functional antagonism. As the first drug to modulate the sphingolipid signaling pathway, it was marketed in 2010 for the treatment of multiple sclerosis (MS). At that time, immunomodulation was widely accepted as the key mechanism of fingolimod’s efficacy in MS. But given the excellent passage of this lipophilic compound into the brain and its massive brain accumulation as well as the abundant expression of S1P receptors on brain cells, it is conceivable that fingolimod also affects brain cells directly. Indeed, a seminal study showed that the protective effect of fingolimod in experimental autoimmune encephalitis (EAE), a murine MS model, is lost in mice lacking the S1P1 receptor on astrocytes, arguing for a specific role of astrocytic S1P signaling in MS. In this review, we discuss the role of sphingolipid mediators and their metabolizing enzymes in neurologic diseases and putative therapeutic strategies arising thereof.
Collapse
Affiliation(s)
- Robert Brunkhorst
- Cerebrovascular Research Group, Department of Neurology, Frankfurt University Hospital Frankfurt am Main, Germany
| | - Rajkumar Vutukuri
- Institute of General Pharmacology and Toxicology, pharmazentrum frankfurt, Goethe University Frankfurt Frankfurt am Main, Germany
| | - Waltraud Pfeilschifter
- Cerebrovascular Research Group, Department of Neurology, Frankfurt University Hospital Frankfurt am Main, Germany
| |
Collapse
|
182
|
Polyglutamine-expanded ataxin-3 impairs long-term depression in Purkinje neurons of SCA3 transgenic mouse by inhibiting HAT and impairing histone acetylation. Brain Res 2014; 1583:220-9. [PMID: 25139423 DOI: 10.1016/j.brainres.2014.08.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 06/11/2014] [Accepted: 08/07/2014] [Indexed: 01/13/2023]
Abstract
Our previous study using a transgenic mouse model of spinocerebellar ataxia type 3 (SCA3) reported that disease-causing ataxin-3-Q79 caused cerebellar malfunction by inducing transcriptional downregulation. Long-term depression (LTD) of parallel fiber-Purkinje neuron glutamatergic transmission is believed to be a cellular mechanism for motor learning and motor coordination in the cerebellum. Downregulated mRNA expression of calcineurin B, IP3-R1, myosin Va and PLC β4, which are required for the induction of cerebellar LTD, led to an impairment of LTD induction in Purkinje neurons of SCA3 transgenic mouse. Our study suggested that ataxin-3-Q79 caused hypoacetylation of cerebellar histone H3 or H4 by inhibiting the activity of histone acetyltransferase (HAT) without affecting the activity of histone deacetylase (HDAC). Consistent with the hypothesis that hypoacetylated H3 or H4 histone associated with promoter regions of downregulated genes is the molecular mechanism underlying ataxin-3-Q79-induced transcriptional repression, chromatin immunoprecipitation-quantitative real-time PCR analysis showed hypoacetylation of H3 or H4 histone associated with the proximal promoter of downregulated calcineurin B, IP3-R1, myosin Va or PLC β4 gene in the cerebellum of SCA3 mouse. HDAC inhibitor sodium butyrate reversed ataxin-3-Q79-induced hypoacetylation of histone H3 or H4 associated with the proximal promoter of calcineurin B, IP3-R1, myosin Va or PLC β4 gene. Sodium butyrate also prevented ataxin-3-Q79-induced impairment of LTD induction in Purkinje neurons of SCA3 mice. Our results suggest that polyglutamine-expanded ataxin-3-Q79 impairs HAT activity, leading to histone hypoacetylation, downregulated expression of cerebellar genes required for LTD induction and impaired induction of cerebellar LTD in the SCA3 transgenic mouse.
Collapse
|
183
|
Fortress AM, Kim J, Poole RL, Gould TJ, Frick KM. 17β-Estradiol regulates histone alterations associated with memory consolidation and increases Bdnf promoter acetylation in middle-aged female mice. ACTA ACUST UNITED AC 2014; 21:457-67. [PMID: 25128537 PMCID: PMC4138358 DOI: 10.1101/lm.034033.113] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Histone acetylation is essential for hippocampal memory formation in young adult rodents. Although dysfunctional histone acetylation has been associated with age-related memory decline in male rodents, little is known about whether histone acetylation is altered by aging in female rodents. In young female mice, the ability of 17β-estradiol (E2) to enhance object recognition memory consolidation requires histone H3 acetylation in the dorsal hippocampus. However, the extent to which histone acetylation is regulated by E2 in middle-aged females is unknown. The mnemonic benefits of E2 in aging females appear to be greatest in middle age, and so pinpointing the molecular mechanisms through which E2 enhances memory at this age could lead to the development of safer and more effective treatments for maintaining memory function without the side effects of current therapies. Here, we show that dorsal hippocampal infusion of E2 rapidly enhanced object recognition and spatial memory, and increased histone H3 acetylation in the dorsal hippocampus, while also significantly reducing levels of histone deacetylase (HDAC2 and HDAC3) proteins. E2 specifically increased histone H3 acetylation at Bdnf promoters pII and pIV in the dorsal hippocampus of both young and middle-aged mice, despite age-related decreases in pI and pIV acetylation. Furthermore, levels of mature BDNF and pro-BDNF proteins in the dorsal hippocampus were increased by E2 in middle-aged females. Together, these data suggest that the middle-aged female dorsal hippocampus remains epigenetically responsive to E2, and that E2 may enhance memory in middle-aged females via epigenetic regulation of Bdnf.
Collapse
Affiliation(s)
- Ashley M Fortress
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| | - Jaekyoon Kim
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| | - Rachel L Poole
- Department of Psychology, Temple University, Philadelphia, Pennsylvania 19122, USA
| | - Thomas J Gould
- Department of Psychology, Temple University, Philadelphia, Pennsylvania 19122, USA
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| |
Collapse
|
184
|
Class-IIa Histone Deacetylase Inhibition Promotes the Growth of Neural Processes and Protects Them Against Neurotoxic Insult. Mol Neurobiol 2014; 51:1432-42. [DOI: 10.1007/s12035-014-8820-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 07/15/2014] [Indexed: 11/25/2022]
|
185
|
Stilling RM, Rönicke R, Benito E, Urbanke H, Capece V, Burkhardt S, Bahari-Javan S, Barth J, Sananbenesi F, Schütz AL, Dyczkowski J, Martinez-Hernandez A, Kerimoglu C, Dent SYR, Bonn S, Reymann KG, Fischer A. K-Lysine acetyltransferase 2a regulates a hippocampal gene expression network linked to memory formation. EMBO J 2014; 33:1912-27. [PMID: 25024434 DOI: 10.15252/embj.201487870] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Neuronal histone acetylation has been linked to memory consolidation, and targeting histone acetylation has emerged as a promising therapeutic strategy for neuropsychiatric diseases. However, the role of histone-modifying enzymes in the adult brain is still far from being understood. Here we use RNA sequencing to screen the levels of all known histone acetyltransferases (HATs) in the hippocampal CA1 region and find that K-acetyltransferase 2a (Kat2a)--a HAT that has not been studied for its role in memory function so far--shows highest expression. Mice that lack Kat2a show impaired hippocampal synaptic plasticity and long-term memory consolidation. We furthermore show that Kat2a regulates a highly interconnected hippocampal gene expression network linked to neuroactive receptor signaling via a mechanism that involves nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). In conclusion, our data establish Kat2a as a novel and essential regulator of hippocampal memory consolidation.
Collapse
Affiliation(s)
- Roman M Stilling
- Department of Psychiatry and Psychotherapy, University Medical Center, Göttingen, Germany
| | - Raik Rönicke
- Research group for Pathophysiology in Dementia, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Eva Benito
- Research group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Hendrik Urbanke
- Research group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Vincenzo Capece
- Research group for Computational Analysis of Biological Networks, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Susanne Burkhardt
- Research group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Sanaz Bahari-Javan
- Research group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Jonas Barth
- Research group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Farahnaz Sananbenesi
- Department of Psychiatry and Psychotherapy, University Medical Center, Göttingen, Germany
| | - Anna L Schütz
- Research group for Computational Analysis of Biological Networks, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Jerzy Dyczkowski
- Research group for Computational Analysis of Biological Networks, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Ana Martinez-Hernandez
- Research group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Cemil Kerimoglu
- Department of Psychiatry and Psychotherapy, University Medical Center, Göttingen, Germany
| | - Sharon Y R Dent
- MD Anderson Cancer Center, University of Texas, Smithville, TX, USA
| | - Stefan Bonn
- Research group for Computational Analysis of Biological Networks, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Klaus G Reymann
- Research group for Pathophysiology in Dementia, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Andre Fischer
- Department of Psychiatry and Psychotherapy, University Medical Center, Göttingen, Germany Research group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| |
Collapse
|
186
|
Zheng X, Zhang X, Kang A, Ran C, Wang G, Hao H. Thinking outside the brain for cognitive improvement: Is peripheral immunomodulation on the way? Neuropharmacology 2014; 96:94-104. [PMID: 24978103 DOI: 10.1016/j.neuropharm.2014.06.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 06/16/2014] [Accepted: 06/19/2014] [Indexed: 02/07/2023]
Abstract
Cognitive impairment is a devastating condition commonly observed with normal aging and neurodegenerative disorders such as Alzheimer's Disease (AD). Although major efforts to prevent or slow down cognitive decline are largely focused within the central nervous system (CNS), it has become clear that signals from the systemic milieu are closely associated with the dysfunctional brain. In particular, the bidirectional crosstalk between the CNS and peripheral immune system plays a decisive role in shaping neuronal survival and function via neuroimmune, neuroendocrinal and bioenergetic mechanisms. Importantly, it is emerging that some neuroprotective and cognition-strengthening drugs may work by targeting the brain-periphery interactions, which could be intriguingly achieved without entering the CNS. We describe here how recent advances in dissecting cognitive deficits from a systems-perspective have contributed to a non-neurocentric understanding of its pathogenesis and treatment strategy. We also discuss the therapeutic and diagnostic implications of these exciting progresses and consider some key issues in the clinical translation. This article is part of a Special Issue entitled 'Neuroimmunology and Synaptic Function'.
Collapse
Affiliation(s)
- Xiao Zheng
- Nanjing University of Chinese Medicine Affiliated Hospital, Nanjing 210029, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Xueli Zhang
- Zhong Da Hospital, Southeast University, Nanjing 210009, China
| | - An Kang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chongzhao Ran
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Boston 02129, United States
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
187
|
Bahari-Javan S, Sananbenesi F, Fischer A. Histone-acetylation: a link between Alzheimer's disease and post-traumatic stress disorder? Front Neurosci 2014; 8:160. [PMID: 25009454 PMCID: PMC4067694 DOI: 10.3389/fnins.2014.00160] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 05/26/2014] [Indexed: 11/13/2022] Open
Abstract
The orchestration of gene-expression programs is essential for cellular homeostasis. Epigenetic processes provide to the cell a key mechanism that allows the regulation of gene-expression networks in response to environmental stimuli. Recently epigenetic mechanisms such as histone-modifications have been implicated with cognitive function and altered epigenome plasticity has been linked to the pathogenesis of neurodegenerative and neuropsychiatric diseases. Thus, key regulators of epigenetic gene-expression have emerged as novel drug targets for brain diseases. Numerous recent review articles discuss in detail the current findings of epigenetic processes in brain diseases. The aim of this article is not to give yet another comprehensive overview of the field but to specifically address the question why the same epigenetic therapies that target histone-acetylation may be suitable to treat seemingly different diseases such as Alzheimer's disease and post-traumatic stress disorder.
Collapse
Affiliation(s)
- Sanaz Bahari-Javan
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen Göttingen, Germany ; Research Group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen Germany
| | - Farahnaz Sananbenesi
- Research Group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen Germany
| | - Andre Fischer
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen Göttingen, Germany ; Research Group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen Germany
| |
Collapse
|
188
|
Sellmann C, Villarín Pildaín L, Schmitt A, Leonardi-Essmann F, Durrenberger PF, Spanagel R, Arzberger T, Kretzschmar H, Zink M, Gruber O, Herrera-Marschitz M, Reynolds R, Falkai P, Gebicke-Haerter PJ, Matthäus F. Gene expression in superior temporal cortex of schizophrenia patients. Eur Arch Psychiatry Clin Neurosci 2014; 264:297-309. [PMID: 24287731 DOI: 10.1007/s00406-013-0473-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 11/12/2013] [Indexed: 11/24/2022]
Abstract
We investigated gene expression pattern obtained from microarray data of 10 schizophrenia patients and 10 control subjects. Brain tissue samples were obtained postmortem; thus, the different ages of the patients at death also allowed a study of the dynamic behavior of the expression patterns over a time frame of many years. We used statistical tests and dimensionality reduction methods to characterize the subset of genes differentially expressed in the two groups. A set of 10 genes were significantly downregulated, and a larger set of 40 genes were upregulated in the schizophrenia patients. Interestingly, the set of upregulated genes includes a large number of genes associated with gene transcription (zinc finger proteins and histone methylation) and apoptosis. We furthermore identified genes with a significant trend correlating with age in the control (MLL3) or the schizophrenia group (SOX5, CTRL). Assessments of correlations of other genes with the disorder (RRM1) or with the duration of medication could not be resolved, because all patients were medicated. This hypothesis-free approach uncovered a series of genes differentially expressed in schizophrenia that belong to a number of distinct cell functions, such as apoptosis, transcriptional regulation, cell motility, energy metabolism and hypoxia.
Collapse
Affiliation(s)
- C Sellmann
- Institute for Pharmacy and Molecular Biotechnology, University of Heidelberg, Im Neuenheimer Feld 364, 69120, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
189
|
Active, phosphorylated fingolimod inhibits histone deacetylases and facilitates fear extinction memory. Nat Neurosci 2014; 17:971-80. [PMID: 24859201 DOI: 10.1038/nn.3728] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 04/25/2014] [Indexed: 02/08/2023]
Abstract
FTY720 (fingolimod), an FDA-approved drug for treatment of multiple sclerosis, has beneficial effects in the CNS that are not yet well understood, independent of its effects on immune cell trafficking. We show that FTY720 enters the nucleus, where it is phosphorylated by sphingosine kinase 2 (SphK2), and that nuclear FTY720-P binds and inhibits class I histone deacetylases (HDACs), enhancing specific histone acetylations. FTY720 is also phosphorylated in mice and accumulates in the brain, including the hippocampus, inhibits HDACs and enhances histone acetylation and gene expression programs associated with memory and learning, and rescues memory deficits independently of its immunosuppressive actions. Sphk2(-/-) mice have lower levels of hippocampal sphingosine-1-phosphate, an endogenous HDAC inhibitor, and reduced histone acetylation, and display deficits in spatial memory and impaired contextual fear extinction. Thus, sphingosine-1-phosphate and SphK2 play specific roles in memory functions and FTY720 may be a useful adjuvant therapy to facilitate extinction of aversive memories.
Collapse
|
190
|
Levi S, Finazzi D. Neurodegeneration with brain iron accumulation: update on pathogenic mechanisms. Front Pharmacol 2014; 5:99. [PMID: 24847269 PMCID: PMC4019866 DOI: 10.3389/fphar.2014.00099] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/17/2014] [Indexed: 12/21/2022] Open
Abstract
Perturbation of iron distribution is observed in many neurodegenerative disorders, including Alzheimer’s and Parkinson’s disease, but the comprehension of the metal role in the development and progression of such disorders is still very limited. The combination of more powerful brain imaging techniques and faster genomic DNA sequencing procedures has allowed the description of a set of genetic disorders characterized by a constant and often early accumulation of iron in specific brain regions and the identification of the associated genes; these disorders are now collectively included in the category of neurodegeneration with brain iron accumulation (NBIA). So far 10 different genetic forms have been described but this number is likely to increase in short time. Two forms are linked to mutations in genes directly involved in iron metabolism: neuroferritinopathy, associated to mutations in the FTL gene and aceruloplasminemia, where the ceruloplasmin gene product is defective. In the other forms the connection with iron metabolism is not evident at all and the genetic data let infer the involvement of other pathways: Pank2, Pla2G6, C19orf12, COASY, and FA2H genes seem to be related to lipid metabolism and to mitochondria functioning, WDR45 and ATP13A2 genes are implicated in lysosomal and autophagosome activity, while the C2orf37 gene encodes a nucleolar protein of unknown function. There is much hope in the scientific community that the study of the NBIA forms may provide important insight as to the link between brain iron metabolism and neurodegenerative mechanisms and eventually pave the way for new therapeutic avenues also for the more common neurodegenerative disorders. In this work, we will review the most recent findings in the molecular mechanisms underlining the most common forms of NBIA and analyze their possible link with brain iron metabolism.
Collapse
Affiliation(s)
- Sonia Levi
- Proteomic of Iron Metabolism, Vita-Salute San Raffaele University Milano, Italy ; San Raffaele Scientific Institute Milano, Italy
| | - Dario Finazzi
- Department of Molecular and Translational Medicine, University of Brescia Brescia, Italy ; Spedali Civili di Brescia Brescia, Italy
| |
Collapse
|
191
|
Valor LM. Transcription, epigenetics and ameliorative strategies in Huntington's Disease: a genome-wide perspective. Mol Neurobiol 2014; 51:406-23. [PMID: 24788684 PMCID: PMC4309905 DOI: 10.1007/s12035-014-8715-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 04/11/2014] [Indexed: 12/18/2022]
Abstract
Transcriptional dysregulation in Huntington’s disease (HD) is an early event that shapes the brain transcriptome by both the depletion and ectopic activation of gene products that eventually affect survival and neuronal functions. Disruption in the activity of gene expression regulators, such as transcription factors, chromatin-remodeling proteins, and noncoding RNAs, accounts for the expression changes observed in multiple animal and cellular models of HD and in samples from patients. Here, I review the recent advances in the study of HD transcriptional dysregulation and its causes to finally discuss the possible implications in ameliorative strategies from a genome-wide perspective. To date, the use of genome-wide approaches, predominantly based on microarray platforms, has been successful in providing an extensive catalog of differentially regulated genes, including biomarkers aimed at monitoring the progress of the pathology. Although still incipient, the introduction of combined next-generation sequencing techniques is enhancing our comprehension of the mechanisms underlying altered transcriptional dysregulation in HD by providing the first genomic landscapes associated with epigenetics and the occupancy of transcription factors. In addition, the use of genome-wide approaches is becoming more and more necessary to evaluate the efficacy and safety of ameliorative strategies and to identify novel mechanisms of amelioration that may help in the improvement of current preclinical therapeutics. Finally, the major conclusions obtained from HD transcriptomics studies have the potential to be extrapolated to other neurodegenerative disorders.
Collapse
Affiliation(s)
- Luis M Valor
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas, Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant, 03550, Alicante, Spain,
| |
Collapse
|
192
|
Murphy SP, Lee RJ, McClean ME, Pemberton HE, Uo T, Morrison RS, Bastian C, Baltan S. MS-275, a class I histone deacetylase inhibitor, protects the p53-deficient mouse against ischemic injury. J Neurochem 2014; 129:509-15. [PMID: 24147654 PMCID: PMC8937574 DOI: 10.1111/jnc.12498] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 10/16/2013] [Accepted: 10/17/2013] [Indexed: 01/24/2023]
Abstract
The administration of pan histone deacetylase (HDAC) inhibitors reduces ischemic damage to the CNS, both in vitro and in animal models of stroke, via mechanisms which we are beginning to understand. The acetylation of p53 is regulated by Class I HDACs and, because p53 appears to play a role in ischemic pathology, the purpose of this study was to discover, using an in vitro white matter ischemia model and an in vivo cerebral ischemia model, if neuroprotection mediated by HDAC inhibition depended on p53 expression. Optic nerves were excised from wild-type and p53-deficient mice, and then subjected to oxygen-glucose deprivation in the presence and absence of a specific inhibitor of Class I HDACs (MS-275, entinostat) while compound action potentials were recorded. Furthermore, transient focal ischemia was imposed on wild-type and p53-deficient mice, which were subsequently treated with MS-275. Interestingly, and in both scenarios, the beneficial effects of MS-275 were most pronounced when p53 was absent. These results suggest that modulation of p53 activity is not responsible for MS-275-mediated neuroprotection, and further illustrate how HDAC inhibitors variably influence p53 and associated apoptotic pathways. Optic nerves from wild-type and p53-deficient mice, engineered to express cyan fluorescent protein (CFP) in neuronal mitochondria, were subjected to oxygen-glucose deprivation (OGD) in the presence and absence of a specific inhibitor of Class I histone deacetylases. The protective effect of MS-275 was evidenced by mitochondrial preservation, and this was most pronounced in the absence of p53.
Collapse
Affiliation(s)
- Sean P. Murphy
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| | - Rona J. Lee
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| | - Megan E. McClean
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| | - Heather E. Pemberton
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| | - Takuma Uo
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| | - Richard S. Morrison
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| | - Chinthasagar Bastian
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Selva Baltan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
193
|
Castellano JF, Fletcher BR, Patzke H, Long JM, Sewal A, Kim DH, Kelley-Bell B, Rapp PR. Reassessing the effects of histone deacetylase inhibitors on hippocampal memory and cognitive aging. Hippocampus 2014; 24:1006-16. [PMID: 24753063 DOI: 10.1002/hipo.22286] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2014] [Indexed: 12/12/2022]
Abstract
Converging results link histone acetylation dynamics to hippocampus-dependent memory, including evidence that histone deacetylase inhibitor (HDACi) administration enhances long-term memory. Previously, we demonstrated that aging disrupts the coordinated epigenetic response to recent experience observed in the young adult hippocampus. Here, we extended that work to test the cognitive effects of a novel, brain-penetrant HDACi (EVX001688; EVX) that we confirmed yields robust, relatively long lasting dose-dependent increases in histone acetylation in the hippocampus. In young rats, acute systemic EVX administration, scheduled to yield elevated histone acetylation levels during training in a contextual fear conditioning (CFC) task, had no effect on memory retention at 24 h at any dose examined (10, 30, or 60 mg/kg). Pretraining injection of another HDACi, sodium butyrate, also failed to affect fear memory, and CFC training itself had no influence on hippocampal histone acetylation at 1 hour in mice or two strains of rats. EVX administration before water maze training in young rats yielded a modest effect such that the middle dose produced marginally better 24-h retention than either the low or high dose, but only a small numerical benefit relative to vehicle. Guided by those findings, a final experiment tested the influence of pretraining EVX treatment on age-related spatial memory impairment. The results, revealing no effect on performance, are consistent with the idea that effective procognitive HDACi treatments in aging may require intervention aimed at restoring coordinated epigenetic regulation rather than bulk increases in hippocampal histone acetylation.
Collapse
Affiliation(s)
- James F Castellano
- Neurocognitive Aging Section, National Institute on Aging, Baltimore, Maryland; Graduate Program in Neuroscience, Icahn School of Medicine at Mount Sinai, New York
| | | | | | | | | | | | | | | |
Collapse
|
194
|
Epigenetic Regulation of Memory by Acetylation and Methylation of Chromatin: Implications in Neurological Disorders, Aging, and Addiction. Neuromolecular Med 2014; 17:97-110. [DOI: 10.1007/s12017-014-8306-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 04/15/2014] [Indexed: 12/11/2022]
|
195
|
Wang X, Xu Q, Bey AL, Lee Y, Jiang YH. Transcriptional and functional complexity of Shank3 provides a molecular framework to understand the phenotypic heterogeneity of SHANK3 causing autism and Shank3 mutant mice. Mol Autism 2014; 5:30. [PMID: 25071925 PMCID: PMC4113141 DOI: 10.1186/2040-2392-5-30] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 03/13/2014] [Indexed: 01/24/2023] Open
Abstract
Background Considerable clinical heterogeneity has been well documented amongst individuals with autism spectrum disorders (ASD). However, little is known about the biological mechanisms underlying phenotypic diversity. Genetic studies have established a strong causal relationship between ASD and molecular defects in the SHANK3 gene. Individuals with various defects of SHANK3 display considerable clinical heterogeneity. Different lines of Shank3 mutant mice with deletions of different portions of coding exons have been reported recently. Variable synaptic and behavioral phenotypes have been reported in these mice, which makes the interpretations for these data complicated without the full knowledge of the complexity of the Shank3 transcript structure. Methods We systematically examined alternative splicing and isoform-specific expression of Shank3 across different brain regions and developmental stages by regular RT-PCR, quantitative real time RT-PCR (q-PCR), and western blot. With these techniques, we also investigated the effects of neuronal activity and epigenetic modulation on alternative splicing and isoform-specific expression of Shank3. We explored the localization and influence on dendritic spine development of different Shank3 isoforms in cultured hippocampal neurons by cellular imaging. Results The Shank3 gene displayed an extensive array of mRNA and protein isoforms resulting from the combination of multiple intragenic promoters and extensive alternative splicing of coding exons in the mouse brain. The isoform-specific expression and alternative splicing of Shank3 were brain-region/cell-type specific, developmentally regulated, activity-dependent, and involved epigenetic regulation. Different subcellular distribution and differential effects on dendritic spine morphology were observed for different Shank3 isoforms. Conclusions Our results indicate a complex transcriptional regulation of Shank3 in mouse brains. Our analysis of select Shank3 isoforms in cultured neurons suggests that different Shank3 isoforms have distinct functions. Therefore, the different types of SHANK3 mutations found in patients with ASD and different exonic deletions of Shank3 in mutant mice are predicted to disrupt selective isoforms and result in distinct dysfunctions at the synapse with possible differential effects on behavior. Our comprehensive data on Shank3 transcriptional regulation thus provides an essential molecular framework to understand the phenotypic diversity in SHANK3 causing ASD and Shank3 mutant mice.
Collapse
Affiliation(s)
- Xiaoming Wang
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, 27710 Durham, NC, USA
| | - Qiong Xu
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, 27710 Durham, NC, USA ; Department of Child Health Care, Children's Hospital of Fudan University, 201102 Shanghai, China
| | - Alexandra L Bey
- Department of Neurobiology, Duke University School of Medicine, 27710 Durham, NC, USA
| | - Yoonji Lee
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, 27710 Durham, NC, USA
| | - Yong-Hui Jiang
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, 27710 Durham, NC, USA ; Department of Neurobiology, Duke University School of Medicine, 27710 Durham, NC, USA
| |
Collapse
|
196
|
Abstract
Recent data support the view that epigenetic processes play a role in memory consolidation and help to transmit acquired memories even across generations in a Lamarckian manner. Drugs that target the epigenetic machinery were found to enhance memory function in rodents and ameliorate disease phenotypes in models for brain diseases such as Alzheimer's disease, Chorea Huntington, Depression or Schizophrenia. In this review, I will give an overview on the current knowledge of epigenetic processes in memory function and brain disease with a focus on Morbus Alzheimer as the most common neurodegenerative disease. I will address the question whether an epigenetic therapy could indeed be a suitable therapeutic avenue to treat brain diseases and discuss the necessary steps that should help to take neuroepigenetic research to the next level.
Collapse
Affiliation(s)
- Andre Fischer
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
197
|
Valor LM, Viosca J, Lopez-Atalaya JP, Barco A. Lysine acetyltransferases CBP and p300 as therapeutic targets in cognitive and neurodegenerative disorders. Curr Pharm Des 2014; 19:5051-64. [PMID: 23448461 PMCID: PMC3722569 DOI: 10.2174/13816128113199990382] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 02/18/2013] [Indexed: 01/27/2023]
Abstract
Neuropsychiatric pathologies, including neurodegenerative diseases and neurodevelopmental syndromes, are frequently associated with dysregulation of various essential cellular mechanisms, such as transcription, mitochondrial respiration and protein degradation. In these complex scenarios, it is difficult to pinpoint the specific molecular dysfunction that initiated the pathology or that led to the fatal cascade of events that ends with the death of the neuron. Among the possible original factors, epigenetic dysregulation has attracted special attention. This review focuses on two highly related epigenetic factors that are directly involved in a number of neurological disorders, the lysine acetyltransferases CREB-binding protein (CBP) and E1A-associated protein p300 (p300). We first comment on the role of chromatin acetylation and the enzymes that control it, particularly CBP and p300, in neuronal plasticity and cognition. Next, we describe the involvement of these proteins in intellectual disability and in different neurodegenerative diseases. Finally, we discuss the potential of ameliorative strategies targeting CBP/p300 for the treatment of these disorders.
Collapse
Affiliation(s)
- Luis M Valor
- Instituto de Neurociencias, Av. Santiago Ramon y Cajal s/n. Sant Joan d'Alacant 03550, Alicante, Spain
| | | | | | | |
Collapse
|
198
|
Yoo JYJ, Larouche M, Goldowitz D. The expression of HDAC1 and HDAC2 during cerebellar cortical development. THE CEREBELLUM 2014; 12:534-46. [PMID: 23436026 DOI: 10.1007/s12311-013-0459-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Histone deacetylases (HDACs) are epigenetic regulatory proteins that repress gene transcription by changing DNA conformation. The regulation of gene expression through histone deacetylation is an important mechanism for the development of the central nervous system. Although the disruption of the balance in epigenetic gene regulation has been implicated in many CNS developmental abnormalities and diseases, the expression pattern of HDACs in various cell types in the brain during its maturation process has had limited exploration. Therefore, in this study, we investigate the cell type-specific and developmental stage-specific expression pattern of HDAC1 and HDAC2 in the mouse cerebellum. Our experimental results show that the cerebellar progenitors and glial cells express high levels of HDAC1 and low levels of HDAC2. On the other hand, the post-mitotic migrating neuronal cells of the cerebellar cortex show strong HDAC2 and weak HDAC1 expressions. In more differentiated neuronal cells, including Purkinje cells, granule cells, unipolar brush cells, and GABAergic interneurons, we found a consistent expression pattern, high levels of HDAC2 and low levels of HDAC1. Therefore, our data provide support for the potential important roles of HDAC1 in cell proliferation and HDAC2 in migration and differentiation.
Collapse
Affiliation(s)
- Ji Young Janice Yoo
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, 950 W. 28th Avenue, Vancouver, BC, Canada
| | | | | |
Collapse
|
199
|
Implications of epigenetic modulation for novel treatment approaches in patients with schizophrenia. Neuropharmacology 2014; 77:481-6. [DOI: 10.1016/j.neuropharm.2013.08.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Revised: 08/21/2013] [Accepted: 08/30/2013] [Indexed: 11/21/2022]
|
200
|
Fischer A. Targeting histone-modifications in Alzheimer's disease. What is the evidence that this is a promising therapeutic avenue? Neuropharmacology 2014; 80:95-102. [PMID: 24486385 DOI: 10.1016/j.neuropharm.2014.01.038] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 01/20/2014] [Accepted: 01/21/2014] [Indexed: 01/06/2023]
Abstract
Alzheimer' s disease (AD) is the most common form of dementia causing an increasing emotional and economical burden to our societies. Although much progress has been made regarding the molecular mechanisms that underlie AD pathogenesis effective therapies are not available yet. The emerging field of neuroepigenetics has provided evidence that de-regulation of epigenetic processes play a role in AD. In this article we will critically review the primary research data that led to the hypothesis that targeting histone-modifying enzymes could be used to treat AD pathogenesis and address the question if the field is ready to translate such findings into clinical application.
Collapse
Affiliation(s)
- Andre Fischer
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Grisebachstr. 5, 37077 Göttingen, Germany; Research Group for Epigenetic Mechansims of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, Grisebachstr. 5, 37077 Göttingen, Germany.
| |
Collapse
|