201
|
Kynurenic acid: a new effector of valproate action? Pharmacol Rep 2011; 63:1569-73. [DOI: 10.1016/s1734-1140(11)70723-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Revised: 06/21/2011] [Indexed: 12/31/2022]
|
202
|
Correa F, Mallard C, Nilsson M, Sandberg M. Activated microglia decrease histone acetylation and Nrf2-inducible anti-oxidant defence in astrocytes: restoring effects of inhibitors of HDACs, p38 MAPK and GSK3β. Neurobiol Dis 2011; 44:142-51. [PMID: 21757005 PMCID: PMC3341174 DOI: 10.1016/j.nbd.2011.06.016] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 05/31/2011] [Accepted: 06/23/2011] [Indexed: 12/21/2022] Open
Abstract
Histone deacetylase (HDAC) inhibitors have promising neuroprotective and anti-inflammatory properties although the exact mechanisms are unclear. We have earlier showed that factors from lipopolysaccharide (LPS)-activated microglia can down-regulate the astroglial nuclear factor-erythroid 2-related factor 2 (Nrf2)-inducible anti-oxidant defence. Here we have evaluated whether histone modification and activation of GSK3β are involved in these negative effects of microglia. Microglia were cultured for 24 h in serum-free culture medium to achieve microglia-conditioned medium from non-activated cells (MCM(0)) or activated with 10 ng/mL of LPS to produce MCM(10). Astrocyte-rich cultures treated with MCM(10) showed a time-dependent (0-72 h) increase in astroglial HDAC activity that correlated with lower levels of acetylation of histones H3 and H4 and decreased levels of the transcription factor Nrf2 and γ-glutamyl cysteine ligase modulatory subunit (γGCL-M) protein levels. The HDAC inhibitors valproic acid (VPA) and trichostatin-A (TSA) elevated the histone acetylation levels, restored the Nrf2-inducible anti-oxidant defence and conferred protection from oxidative stress-induced (H(2)O(2)) death in astrocyte-rich cultures exposed to MCM(10). Inhibitors of GSK3β (lithium) and p38 MAPK (SB203580) signaling pathways restored the depressed histone acetylation and Nrf2-related transcription whereas an inhibitor of Akt (Ly294002) caused a further decrease in Nrf2-related transcription. In conclusion, the study shows that well tolerated drugs such as VPA and lithium can restore an inflammatory induced depression in the Nrf2-inducible antioxidant defence, possibly via normalised histone acetylation levels.
Collapse
Affiliation(s)
- Fernando Correa
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Sweden.
| | | | | | | |
Collapse
|
203
|
Circulating levels of GDNF in bipolar disorder. Neurosci Lett 2011; 502:103-6. [DOI: 10.1016/j.neulet.2011.07.031] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 07/19/2011] [Accepted: 07/19/2011] [Indexed: 11/18/2022]
|
204
|
Villalba RM, Smith Y. Neuroglial plasticity at striatal glutamatergic synapses in Parkinson's disease. Front Syst Neurosci 2011; 5:68. [PMID: 21897810 PMCID: PMC3159891 DOI: 10.3389/fnsys.2011.00068] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Accepted: 08/02/2011] [Indexed: 01/08/2023] Open
Abstract
Striatal dopamine denervation is the pathological hallmark of Parkinson's disease (PD). Another major pathological change described in animal models and PD patients is a significant reduction in the density of dendritic spines on medium spiny striatal projection neurons. Simultaneously, the ultrastructural features of the neuronal synaptic elements at the remaining corticostriatal and thalamostriatal glutamatergic axo-spinous synapses undergo complex ultrastructural remodeling consistent with increased synaptic activity (Villalba and Smith, 2011). The concept of tripartite synapses (TS) was introduced a decade ago, according to which astrocytes process and exchange information with neuronal synaptic elements at glutamatergic synapses (Araque et al., 1999a). Although there has been compelling evidence that astrocytes are integral functional elements of tripartite glutamatergic synaptic complexes in the cerebral cortex and hippocampus, their exact functional role, degree of plasticity and preponderance in other CNS regions remain poorly understood. In this review, we discuss our recent findings showing that neuronal elements at cortical and thalamic glutamatergic synapses undergo significant plastic changes in the striatum of MPTP-treated parkinsonian monkeys. We also present new ultrastructural data that demonstrate a significant expansion of the astrocytic coverage of striatal TS synapses in the parkinsonian state, providing further evidence for ultrastructural compensatory changes that affect both neuronal and glial elements at TS. Together with our limited understanding of the mechanisms by which astrocytes respond to changes in neuronal activity and extracellular transmitter homeostasis, the role of both neuronal and glial components of excitatory synapses must be considered, if one hopes to take advantage of glia–neuronal communication knowledge to better understand the pathophysiology of striatal processing in parkinsonism, and develop new PD therapeutics.
Collapse
Affiliation(s)
- Rosa M Villalba
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University Atlanta, GA, USA
| | | |
Collapse
|
205
|
Protective effects of valproic acid on the nigrostriatal dopamine system in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson's disease. Neuroscience 2011; 194:189-94. [PMID: 21846494 DOI: 10.1016/j.neuroscience.2011.08.010] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 07/29/2011] [Accepted: 08/03/2011] [Indexed: 11/22/2022]
Abstract
The use of animal models (including the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine [MPTP] mouse model) to mimic dopaminergic (DAergic) cell loss and striatal dopamine (DA) depletion, as seen in Parkinson's disease (PD), has implicated a multitude of factors that might be associated with DAergic cell death in PD including excitotoxicity, inflammation, and oxidative stress. All of these factors have been shown to be reduced by administration of histone deacetylase (HDAC) inhibitors (HDACis) resulting in some degree of neuroprotection in various models of neurodegenerative disease including in Huntington's disease and amyotrophic lateral sclerosis. However, there is limited information of effects of HDACis in PD models. We have previously shown HDACis to be partially protective against 1-methyl-4-phenylpyridinium (MPP(+))-mediated cell loss in vitro. The present study was conducted to extend these findings to an in vivo PD model. The HDACi valproic acid (VPA) was co-administered with MPTP for 5 days to male FVBn mice and continued for an additional 2 weeks, throughout the period of active neurodegeneration associated with MPTP-mediated DAergic cell loss. VPA was able to partially prevent striatal dopamine depletion and almost completely protect against substantia nigra DAergic cell loss. These results suggest that VPA may be a potential disease-modifying therapy for PD.
Collapse
|
206
|
Yu Z, Ono C, Kim HB, Komatsu H, Tanabe Y, Sakae N, Nakayama KI, Matsuoka H, Sora I, Bunney WE, Tomita H. Four mood stabilizers commonly induce FEZ1 expression in human astrocytes. Bipolar Disord 2011; 13:486-99. [PMID: 22017218 DOI: 10.1111/j.1399-5618.2011.00946.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Mood stabilizers influence the morphology, chemotaxis, and survival of neurons, which are considered to be related to the mood-stabilizing effects of these drugs. Although previous studies suggest glial abnormalities in patients with bipolar disorder and an effect of mood stabilizers on certain genes in astrocytes, less is known about the effects of mood stabilizers in astrocytes than in neurons. The present study identifies a common underlying response to mood stabilizers in astrocytes. METHODS Human astrocyte-derived cells (U-87 MG) were treated with the four most commonly used mood stabilizers (lithium, valproic acid, carbamazepine, and lamotrigine) and subjected to microarray gene expression analyses. The most prominently regulated genes were validated by qRT-PCR and western blot analysis. The intercellular localization of one of these regulated genes, fasciculation and elongation protein zeta 1 (FEZ1), was evaluated by immunofluorescence staining. RESULTS The microarray data indicated that FEZ1 was the only gene commonly induced by the four mood stabilizers in human astrocyte-derived cells. An independent experiment confirmed astrocytic FEZ1 induction at both the transcript and protein levels following mood stabilizer treatments. FEZ1 localized to the cytoplasm of transformed and primary astrocytes from the human adult brain. CONCLUSIONS Our data suggest that FEZ1 may play important roles in human astrocytes, and that mood stabilizers might exert their cytoprotective and mood-stabilizing effects by inducing FEZ1 expression in astrocytes.
Collapse
Affiliation(s)
- Zhiqian Yu
- Department of Biological Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
207
|
Schosser A, Butler AW, Ising M, Perroud N, Uher R, Ng MY, Cohen-Woods S, Craddock N, Owen MJ, Korszun A, Jones L, Jones I, Gill M, Rice JP, Maier W, Mors O, Rietschel M, Lucae S, Binder EB, Preisig M, Perry J, Tozzi F, Muglia P, Aitchison KJ, Breen G, Craig IW, Farmer AE, Müller-Myhsok B, McGuffin P, Lewis CM. Genomewide association scan of suicidal thoughts and behaviour in major depression. PLoS One 2011; 6:e20690. [PMID: 21750702 PMCID: PMC3130038 DOI: 10.1371/journal.pone.0020690] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Accepted: 05/07/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Suicidal behaviour can be conceptualised as a continuum from suicidal ideation, to suicidal attempts to completed suicide. In this study we identify genes contributing to suicidal behaviour in the depression study RADIANT. METHODOLOGY/PRINCIPAL FINDINGS A quantitative suicidality score was composed of two items from the SCAN interview. In addition, the 251 depression cases with a history of serious suicide attempts were classified to form a discrete trait. The quantitative trait was correlated with younger onset of depression and number of episodes of depression, but not with gender. A genome-wide association study of 2,023 depression cases was performed to identify genes that may contribute to suicidal behaviour. Two Munich depression studies were used as replication cohorts to test the most strongly associated SNPs. No SNP was associated at genome-wide significance level. For the quantitative trait, evidence of association was detected at GFRA1, a receptor for the neurotrophin GDRA (p = 2e-06). For the discrete trait of suicide attempt, SNPs in KIAA1244 and RGS18 attained p-values of <5e-6. None of these SNPs showed evidence for replication in the additional cohorts tested. Candidate gene analysis provided some support for a polymorphism in NTRK2, which was previously associated with suicidality. CONCLUSIONS/SIGNIFICANCE This study provides a genome-wide assessment of possible genetic contribution to suicidal behaviour in depression but indicates a genetic architecture of multiple genes with small effects. Large cohorts will be required to dissect this further.
Collapse
Affiliation(s)
- Alexandra Schosser
- MRC Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, United Kingdom
- Department of Psychiatry and Psychotherapy, Medical University Vienna, Vienna, Austria
| | - Amy W. Butler
- MRC Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, United Kingdom
- Department of Psychiatry, University of Hong Kong, Hong Kong, Special Administrative Region, China
| | - Marcus Ising
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Nader Perroud
- MRC Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, United Kingdom
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Rudolf Uher
- MRC Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Mandy Y. Ng
- MRC Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Sarah Cohen-Woods
- MRC Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Nick Craddock
- MRC Centre for Neuropsychiatric Genetics and Genomics, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Michael J. Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Ania Korszun
- Barts and The London Medical School, Queen Mary University of London, London, United Kingdom
| | - Lisa Jones
- Department of Psychiatry, Neuropharmacology and Neurobiology Section, University of Birmingham, Birmingham, United Kingdom
| | - Ian Jones
- MRC Centre for Neuropsychiatric Genetics and Genomics, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Michael Gill
- Department of Psychiatry, Trinity Centre for Health Science, Dublin, Ireland
| | - John P. Rice
- Department of Psychiatry, Washington University, St. Louis, Missouri, United States of America
| | - Wolfgang Maier
- Department of Psychiatry, University of Bonn, Bonn, Germany
| | - Ole Mors
- Centre for Psychiatric Research, Aarhus University Hospital, Risskov, Denmark
| | - Marcella Rietschel
- Division of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Mannheim, Germany
| | | | | | - Martin Preisig
- University Hospital Center and University of Lausanne, Lausanne, Switzerland
| | - Julia Perry
- GlaxoSmithKline Research & Development, Stockley Park, United Kingdom
| | | | - Pierandrea Muglia
- GlaxoSmithKline Research & Development, Verona, Italy
- Department of Psychiatry, University of Toronto, Toronto, Canada
- NeuroSearch A/S, Ballerup, Denmark
| | - Katherine J. Aitchison
- MRC Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Gerome Breen
- MRC Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, United Kingdom
- NIHR Biomedical Research Centre for Mental Health, South London and Maudsley NHS Foundation Trust and Institute of Psychiatry, King's College London, London, United Kingdom
| | - Ian W. Craig
- MRC Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Anne E. Farmer
- MRC Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, United Kingdom
| | | | - Peter McGuffin
- MRC Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Cathryn M. Lewis
- MRC Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, United Kingdom
| |
Collapse
|
208
|
Granulocyte macrophage-colony stimulating factor protects against substantia nigra dopaminergic cell loss in an environmental toxin model of Parkinson's disease. Neurobiol Dis 2011; 43:99-112. [DOI: 10.1016/j.nbd.2011.02.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 01/26/2011] [Accepted: 02/27/2011] [Indexed: 12/21/2022] Open
|
209
|
Marinova Z, Leng Y, Leeds P, Chuang DM. Histone deacetylase inhibition alters histone methylation associated with heat shock protein 70 promoter modifications in astrocytes and neurons. Neuropharmacology 2011; 60:1109-15. [PMID: 20888352 PMCID: PMC3036778 DOI: 10.1016/j.neuropharm.2010.09.022] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Revised: 09/21/2010] [Accepted: 09/22/2010] [Indexed: 01/05/2023]
Abstract
The mood-stabilizing and anticonvulsant drug valproic acid (VPA) inhibits histone deacetylases (HDACs). The aim of the present study was to determine the effect of HDAC inhibition on overall and target gene promoter-associated histone methylation in rat cortical neurons and astrocytes. We found that VPA and other HDAC inhibitors, including sodium butyrate (SB), trichostatin A (TSA), and the Class I HDAC inhibitors MS-275 and apicidin all increased levels of histone 3 lysine 4 dimethylation and trimethylation (H3K4Me2 and H3K4Me3); these processes are linked to transcriptional activation in rat cortical neurons and astrocytes. VPA, SB, TSA, MS-275, and apicidin also upregulated levels of the neuroprotective heat shock protein 70 (HSP70) in rat astrocytes. Moreover, Class I HDAC inhibition by VPA and MS-275 increased H3K4Me2 levels at the HSP70 promoter in astrocytes and neurons. We also found that VPA treatment facilitated the recruitment of acetyltransferase p300 to the HSP70 promoter and that p300 interacted with the transcription factor NF-Y in astrocytes. Taken together, the results suggest that Class I HDAC inhibition is key to upregulating overall and gene-specific H3K4 methylation in primary neuronal and astrocyte cultures. In addition, VPA-induced activation of the HSP70 promoter in astrocytes appears to involve an increase in H3K4Me2 levels and recruitment of p300. This article is part of a Special Issue entitled 'Trends in neuropharmacology: in memory of Erminio Costa'.
Collapse
Affiliation(s)
- Zoya Marinova
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892-1363, USA
| | | | | | | |
Collapse
|
210
|
Hu JP, Xie JW, Wang CY, Wang T, Wang X, Wang SL, Teng WP, Wang ZY. Valproate reduces tau phosphorylation via cyclin-dependent kinase 5 and glycogen synthase kinase 3 signaling pathways. Brain Res Bull 2011; 85:194-200. [DOI: 10.1016/j.brainresbull.2011.03.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 03/03/2011] [Accepted: 03/11/2011] [Indexed: 01/30/2023]
|
211
|
Valproate reduces CHOP levels and preserves oligodendrocytes and axons after spinal cord injury. Neuroscience 2011; 178:33-44. [DOI: 10.1016/j.neuroscience.2011.01.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Revised: 01/05/2011] [Accepted: 01/06/2011] [Indexed: 01/12/2023]
|
212
|
Germaná C, Kempton MJ, Sarnicola A, Christodoulou T, Haldane M, Hadjulis M, Girardi P, Tatarelli R, Frangou S. The effects of lithium and anticonvulsants on brain structure in bipolar disorder. Acta Psychiatr Scand 2010; 122:481-487. [PMID: 20560901 DOI: 10.1111/j.1600-0447.2010.01582.x] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To investigate the effect of lithium, anticonvulsants and antipsychotics on brain structure in bipolar disorder (BD). METHOD A cross-sectional structural brain magnetic resonance imaging study of 74 remitted patients with BD, aged 18-65, who were receiving long-term prophylactic treatment with lithium or anticonvulsants or antipsychotics. Global and regional grey matter, white matter, and cerebrospinal fluid volumes were compared between treatment groups. RESULTS Grey matter in the subgenual anterior cingulate gyrus on the right (extending into the hypothalamus) and in the postcentral gyrus, the hippocampus/amygdale complex and the insula on the left was greater in BD patients on lithium treatment compared to all other treatment groups. CONCLUSION Lithium treatment in BD has a significant effect on brain structure particularly in limbic/paralimbic regions associated with emotional processing.
Collapse
Affiliation(s)
- C Germaná
- Institute of Psychiatry King's College London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
213
|
Yokoyama H, Uchida H, Kuroiwa H, Kasahara J, Araki T. Role of glial cells in neurotoxin-induced animal models of Parkinson's disease. Neurol Sci 2010; 32:1-7. [PMID: 21107876 DOI: 10.1007/s10072-010-0424-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Accepted: 09/08/2010] [Indexed: 11/28/2022]
Abstract
Dopaminergic neurons are selectively vulnerable to oxidative stress and inflammatory attack. The neuronal cell loss in the substantia nigra is associated with a glial response composed markedly of activated microglia and, to a lesser extent, of reactive astrocytes although these glial responses may be the source of neurotrophic factors and can protect against oxidative stress such as reactive oxygen species and reactive nitrogen species. However, the glial response can also mediate a variety of deleterious events related to the production of pro-inflammatory, pro-oxidant reactive species, prostaglandins, cytokines, and so on. In this review, we discuss the possible protective and deleterious effects of glial cells in the neurodegenerative diseases and examine how these factors may contribute to the pathogenesis of Parkinson's disease. This review suggests that further investigation concerning glial reaction in Parkinson's disease may lead to disease-modifying therapeutic approaches and may contribute to the pathogenesis of this disease.
Collapse
Affiliation(s)
- Hironori Yokoyama
- Department of Neurobiology and Therapeutics, Graduate School and Faculty of Pharmaceutical Sciences, The University of Tokushima, 1-78, Sho-machi, Tokushima, 770-8505, Japan
| | | | | | | | | |
Collapse
|
214
|
Sidoryk-Wegrzynowicz M, Wegrzynowicz M, Lee E, Bowman AB, Aschner M. Role of astrocytes in brain function and disease. Toxicol Pathol 2010; 39:115-23. [PMID: 21075920 DOI: 10.1177/0192623310385254] [Citation(s) in RCA: 169] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Astrocytes assume multiple roles in maintaining an optimally suited milieu for neuronal function. Select astrocytic functions include the maintenance of redox potential, the production of trophic factors, the regulation of neurotransmitter and ion concentrations, and the removal of toxins and debris from the cerebrospinal fluid (CSF). Impairments in these and other functions, as well as physiological reactions of astrocytes to injury, can trigger or exacerbate neuronal dysfunction. This review addresses select metabolic interactions between neurons and astrocytes and emphasizes the role of astrocytes in mediating and amplifying the progression of several neurodegenerative disorders, such as Parkinson's disease (PD), hepatic encephalopathy (HE), hyperammonemia (HA), Alzheimer's disease (AD), and ischemia.
Collapse
|
215
|
Hamby ME, Sofroniew MV. Reactive astrocytes as therapeutic targets for CNS disorders. Neurotherapeutics 2010; 7:494-506. [PMID: 20880511 PMCID: PMC2952540 DOI: 10.1016/j.nurt.2010.07.003] [Citation(s) in RCA: 262] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2010] [Revised: 07/19/2010] [Accepted: 07/20/2010] [Indexed: 12/30/2022] Open
Abstract
Reactive astrogliosis has long been recognized as a ubiquitous feature of CNS pathologies. Although its roles in CNS pathology are only beginning to be defined, genetic tools are enabling molecular dissection of the functions and mechanisms of reactive astrogliosis in vivo. It is now clear that reactive astrogliosis is not simply an all-or-nothing phenomenon but, rather, is a finely gradated continuum of molecular, cellular, and functional changes that range from subtle alterations in gene expression to scar formation. These changes can exert both beneficial and detrimental effects in a context-dependent manner determined by specific molecular signaling cascades. Dysfunction of either astrocytes or the process of reactive astrogliosis is emerging as an important potential source of mechanisms that might contribute to, or play primary roles in, a host of CNS disorders via loss of normal or gain of abnormal astrocyte activities. A rapidly growing understanding of the mechanisms underlying astrocyte signaling and reactive astrogliosis has the potential to open doors to identifying many molecules that might serve as novel therapeutic targets for a wide range of neurological disorders. This review considers general principles and examines selected examples regarding the potential of targeting specific molecular aspects of reactive astrogliosis for therapeutic manipulations, including regulation of glutamate, reactive oxygen species, and cytokines.
Collapse
Affiliation(s)
- Mary E. Hamby
- grid.19006.3e0000000096326718Department of Neurobiology, David Geffen School of Medicine, University of California, 90095 Los Angeles, California
| | - Michael V. Sofroniew
- grid.19006.3e0000000096326718Department of Neurobiology, David Geffen School of Medicine, University of California, 90095 Los Angeles, California
| |
Collapse
|
216
|
Sotthibundhu A, Phansuwan-Pujito P, Govitrapong P. Melatonin increases proliferation of cultured neural stem cells obtained from adult mouse subventricular zone. J Pineal Res 2010; 49:291-300. [PMID: 20663047 DOI: 10.1111/j.1600-079x.2010.00794.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Melatonin, a circadian rhythm-promoting molecule secreted mainly by the pineal gland, has a variety of biological functions and neuroprotective effects including control of sleep-wake cycle, seasonal reproduction, and body temperature as well as preventing neuronal cell death induced by neurotoxic substances. Melatonin also modulates neural stem cell (NSC) function including proliferation and differentiation in embryonic brain tissue. However, the involvement of melatonin in adult neurogenesis is still not clear. Here, we report that precursor cells from adult mouse subventricular zone (SVZ) of the lateral ventricle, the main neurogenic area of the adult brain, express melatonin receptors. In addition, precursor cells derived from this area treated with melatonin exhibited increased proliferative activity. However, when cells were treated with luzindole, a competitive inhibitor of melatonin receptors, or pertussis toxin, an uncoupler of Gi from adenylate cyclase, melatonin-induced proliferation was reduced. Under these conditions, melatonin induced the differentiation of precursor cells to neuronal cells without an upregulation of the number of glia cells. Because stem cell replacement is thought to play an important therapeutic role in neurodegenerative diseases, melatonin might be beneficial for stimulating endogenous neural stem cells.
Collapse
|
217
|
Selvi BR, Cassel JC, Kundu TK, Boutillier AL. Tuning acetylation levels with HAT activators: Therapeutic strategy in neurodegenerative diseases. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2010; 1799:840-53. [DOI: 10.1016/j.bbagrm.2010.08.012] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Revised: 08/24/2010] [Accepted: 08/27/2010] [Indexed: 10/19/2022]
|
218
|
The ALDH2 and DRD2/ANKK1 genes interacted in bipolar II but not bipolar I disorder. Pharmacogenet Genomics 2010; 20:500-6. [PMID: 20577142 DOI: 10.1097/fpc.0b013e32833caa2b] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
219
|
Molecular and therapeutic potential and toxicity of valproic acid. J Biomed Biotechnol 2010; 2010. [PMID: 20798865 PMCID: PMC2926634 DOI: 10.1155/2010/479364] [Citation(s) in RCA: 310] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Revised: 05/03/2010] [Accepted: 06/06/2010] [Indexed: 12/13/2022] Open
Abstract
Valproic acid (VPA), a branched short-chain fatty acid, is widely used as an antiepileptic drug and a mood stabilizer. Antiepileptic properties have been attributed to inhibition of Gamma Amino Butyrate (GABA) transaminobutyrate and of ion channels. VPA was recently classified among the Histone Deacetylase Inhibitors, acting directly at the level of gene transcription by inhibiting histone deacetylation and making transcription sites more accessible. VPA is a widely used drug, particularly for children suffering from epilepsy. Due to the increasing number of clinical trials involving VPA, and interesting results obtained, this molecule will be implicated in an increasing number of therapies. However side effects of VPA are substantially described in the literature whereas they are poorly discussed in articles focusing on its therapeutic use. This paper aims to give an overview of the different clinical-trials involving VPA and its side effects encountered during treatment as well as its molecular properties.
Collapse
|
220
|
Chung YC, Ko HW, Bok E, Park ES, Huh SH, Nam JH, Jin BK. The role of neuroinflammation on the pathogenesis of Parkinson's disease. BMB Rep 2010; 43:225-32. [PMID: 20423606 DOI: 10.5483/bmbrep.2010.43.4.225] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Parkinson's Disease (PD) is a common neurodegenerative disease characterized by the progressive degeneration of nigrostriatal dopaminergic (DA) neurons. Although the causative factors of PD remain elusive, many studies on PD animal models or humans suggest that glial activation along with neuroinflammatory processes contribute to the initiation or progression of PD. Additionally, several groups have proposed that dysfunction of the blood-brain barrier (BBB) combined with infiltration of peripheral immune cells play important roles in the degeneration of DA neurons. However, these neuroinflammatory events have only been investigated separately, and the issue of whether these phenomena are neuroprotective or neurotoxic remains controversial. We here review the current knowledge regarding the functions of these neuroinflammatory processes in the brain. Finally, we describe therapeutic strategies for the regulation of neuroinflammation with the goal of improving the symptoms of PD.
Collapse
Affiliation(s)
- Young Cheul Chung
- Department of Biochemistry & Molecular Biology, Neurodegeneration Control Research Center, School of Medicine Kyung Hee University, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
221
|
Kidd SK, Schneider JS. Protection of dopaminergic cells from MPP+-mediated toxicity by histone deacetylase inhibition. Brain Res 2010; 1354:172-8. [PMID: 20654591 DOI: 10.1016/j.brainres.2010.07.041] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Revised: 07/12/2010] [Accepted: 07/14/2010] [Indexed: 01/08/2023]
Abstract
Although the pathophysiological processes involved in dopamine (DA) neuron degeneration in Parkinson's disease (PD) are not completely known, apoptotic cell death has been suggested to be involved and can be modeled in DAergic cell lines using the mitochondrial toxin 1-methyl-4-phenylpyridinium (MPP(+)). Recently, it has been suggested that histone deacetylase inhibitors (HDACIs) may reduce apoptotic cell death in various model systems. However, their utility in interfering with DA cell death remains unclear. The HDACIs sodium butyrate (NaB), valproate (VPA) and suberoylanilide hydroxamic acid (SAHA) were tested for their ability to prevent MPP(+)-mediated cytotoxicity in human derived SK-N-SH and rat derived MES 23.5 cells. All three HDACIs at least partially prevented MPP(+)-mediated apoptotic cell death. The protective effects of these HDACIs coincided with significant increases in histone acetylation. These results suggest that HDACIs may be potentially neuroprotective against DA cell death and should be explored further in animal models of PD.
Collapse
Affiliation(s)
- Sarah K Kidd
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | |
Collapse
|
222
|
Iwata M, Shirayama Y, Ishida H, Hazama GI, Nakagome K. Hippocampal astrocytes are necessary for antidepressant treatment of learned helplessness rats. Hippocampus 2010; 21:877-84. [PMID: 20572198 DOI: 10.1002/hipo.20803] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2010] [Indexed: 12/16/2022]
Abstract
The astrocyte is a major component of the neural network and plays a role in brain function. Previous studies demonstrated changes in the number of astrocytes in depression. In this study, we examined alterations in the number of astrocytes in the learned helplessness (LH) rat, an animal model of depression. The numbers of activated and nonactivated astrocytes in the dentate gyrus (molecular layer, subgranular zone, and hilus), and CA1 and CA3 regions of the hippocampus were significantly increased 2 and 8 days after attainment of LH. Subchronic treatment with imipramine showed a tendency (although not statistically significant) to decrease the LH-induced increment of activated astrocytes in the CA3 region and dentate gyrus. Furthermore, subchronic treatment of naïve rats with imipramine did not alter the numbers of activated and nonactivated astrocytes. However, the antidepressant-like effects of imipramine in the LH paradigm were blocked when fluorocitrate (a reversible inhibitor of astrocyte function) was injected into the dentate gyrus or CA3 region. Injection of fluorocitrate into naive rats failed to induce behavioral deficits in the conditioned avoidance test. These results indicate that astrocytes are responsive to the antidepressant-like effect of imipramine in the dentate gyrus and CA3 region of the hippocampus.
Collapse
Affiliation(s)
- Masaaki Iwata
- Department of Neuropsychiatry, Faculty of Medicine, Tottori University, Yonago, Japan
| | | | | | | | | |
Collapse
|
223
|
Leng Y, Marinova Z, Reis-Fernandes MA, Nau H, Chuang DM. Potent neuroprotective effects of novel structural derivatives of valproic acid: potential roles of HDAC inhibition and HSP70 induction. Neurosci Lett 2010; 476:127-32. [PMID: 20394799 PMCID: PMC3117289 DOI: 10.1016/j.neulet.2010.04.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 04/05/2010] [Accepted: 04/06/2010] [Indexed: 01/05/2023]
Abstract
Emerging evidence suggests that the neuroprotective effects of valproic acid (VPA) occur via inhibition of histone deacetylases (HDACs) and activation of gene expression. This study assessed the ability of four VPA derivatives to cause histone hyperacetylation and protect against glutamate-induced excitotoxicity in cultured neurons. We found that (S)-2-pentyl-4-pentynoic acid (compound III) and (+/-)-2-hexyl-4-pentynoic acid (compound V) were far more potent and robust than VPA in inducing histone hyperacetylation and protecting against glutamate excitotoxicity. Thus, the increase in histone acetylation elicited by compounds III and V was significant at 5microM and reached a maximal increase of 600-700% at 50-100microM, compared with only a 200% increase by VPA at 100microM. The neuroprotective effects of compounds III and V were evident at 10-25microM and reached a complete protection at 50-100microM, while a significant partial protection by VPA was observed at 100microM. These two compounds were also more effective than VPA in increasing HSP70-1a and HSP70-1b mRNA levels. At 50microM, compound V was most robust in increasing HSP-1a mRNA levels, followed by compound III, and then by VPA. HSP-1b mRNA was only significantly upregulated by compounds V and III, but not by VPA or other VPA derivatives under these treatment conditions. Our results suggest that these two VPA derivatives may ultimately be developed into potent neuroprotective drugs in preclinical and clinical studies.
Collapse
Affiliation(s)
- Yan Leng
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health, Bldg. 10, Rm. 3D-38, 10 Center Dr, Bethesda, MD 20892-1363, USA
| | - Zoya Marinova
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health, Bldg. 10, Rm. 3D-38, 10 Center Dr, Bethesda, MD 20892-1363, USA
| | - Marcos A Reis-Fernandes
- Institute for Food Toxicology and Analytical Chemistry, University of Veterinary Medicine, Hannover, Germany
| | - Heinz Nau
- Institute for Food Toxicology and Analytical Chemistry, University of Veterinary Medicine, Hannover, Germany
| | - De-Maw Chuang
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health, Bldg. 10, Rm. 3D-38, 10 Center Dr, Bethesda, MD 20892-1363, USA
| |
Collapse
|
224
|
Abstract
Psychiatric diseases place a tremendous burden on affected individuals, their caregivers, and the health care system. Although evidence exists for a strong inherited component to many of these conditions, dedicated efforts to identify DNA sequence-based causes have not been exceptionally productive, and very few pharmacologic treatment options are clinically available. Many features of psychiatric diseases are consistent with an epigenetic dysregulation, such as discordance of monozygotic twins, late age of onset, parent-of-origin and sex effects, and fluctuating disease course. In recent years, experimental technologies have significantly advanced, permitting indepth studies of the epigenome and its role in maintenance of normal genomic functions, as well as disease etiopathogenesis. Here, we present an epigenetic explanation for many characteristics of psychiatric disease, review the current literature on the epigenetic mechanisms involved in major psychosis, Alzheimer's disease, and autism spectrum disorders, and describe some future directions in the field of psychiatric epigenomics.
Collapse
Affiliation(s)
- Carolyn Ptak
- The Krembil Family Epigenetics Laboratory, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | | |
Collapse
|
225
|
Smith AM, Gibbons HM, Dragunow M. Valproic acid enhances microglial phagocytosis of amyloid-beta(1-42). Neuroscience 2010; 169:505-15. [PMID: 20423723 DOI: 10.1016/j.neuroscience.2010.04.041] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2009] [Revised: 04/01/2010] [Accepted: 04/19/2010] [Indexed: 01/17/2023]
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder manifested by memory loss, confusion and changes in mood. A principal pathology of this debilitating disorder is extracellular deposits of amyloid-beta (Abeta) protein. The "amyloid hypothesis" postulates that a build-up of Abeta protein is responsible for neuronal loss and the ensuing symptoms of AD. One possible mechanism of Abeta clearance, and hence AD therapy, is phagocytosis of Abeta protein by microglial cells. Microglia are the brain's resident immune cells and phagocytosis is one of their innate functions. We are interested in identifying molecules that augment microglial-mediated phagocytosis of Abeta protein. We used the rodent BV-2 microglial cell line which readily phagocytose fluorescent latex beads and synthetic Abeta(1-42) peptide. BV-2 cells treated with the neuroactive drug valproic acid (VPA) showed greatly enhanced phagocytic activity for both latex beads and Abeta. VPA also reduced microglial viability by inducing apoptosis, as previously reported. The relevance of these in vitro results to the treatment of AD is unclear but further investigation into the effects of VPA on the clearance of Abeta through enhanced microglial phagocytosis is warranted.
Collapse
Affiliation(s)
- A M Smith
- Department of Pharmacology, Faculty of Medical and Health Sciences and the National Research Centre for Growth and Development, The University of Auckland, Auckland, New Zealand
| | | | | |
Collapse
|
226
|
Abstract
Anticonvulsant drugs are widely used in psychiatric indications. This includes alcohol and benzodiazepine withdrawal symptoms, panic and anxiety disorders, dementia, schizophrenia, and to some extent personality disorders. Besides pain syndromes, their main domain outside epilepsy, however, is bipolar disorder. Carbamazepine, valproate, and lamotrigine are meanwhile recognized mood stabilizers, but several other antiepileptic drugs have also been tried out with diverging or inconclusive results. Understanding the mechanisms of action and identifying similarities between anticonvulsants effective in bipolar disorder may also enhance our understanding of the underlying pathophysiology of the disorder.
Collapse
Affiliation(s)
- Heinz C R Grunze
- School of Neurology, Neurobiology and Psychiatry, University of Newcastle upon Tyne, UK.
| |
Collapse
|
227
|
Song C, Kanthasamy A, Anantharam V, Sun F, Kanthasamy AG. Environmental neurotoxic pesticide increases histone acetylation to promote apoptosis in dopaminergic neuronal cells: relevance to epigenetic mechanisms of neurodegeneration. Mol Pharmacol 2010; 77:621-32. [PMID: 20097775 PMCID: PMC2847769 DOI: 10.1124/mol.109.062174] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Accepted: 01/19/2010] [Indexed: 11/22/2022] Open
Abstract
Pesticide exposure has been implicated in the etiopathogenesis of Parkinson's disease (PD); in particular, the organochlorine insecticide dieldrin is believed to be associated with PD. Emerging evidence indicates that histone modifications play a critical role in cell death. In this study, we examined the effects of dieldrin treatment on histone acetylation and its role in dieldrin-induced apoptotic cell death in dopaminergic neuronal cells. In mesencephalic dopaminergic neuronal cells, dieldrin induced a time-dependent increase in the acetylation of core histones H3 and H4. Histone acetylation occurred within 10 min of dieldrin exposure indicating that acetylation is an early event in dieldrin neurotoxicity. The hyperacetylation was attributed to dieldrin-induced proteasomal dysfunction, resulting in accumulation of a key histone acetyltransferase (HAT), cAMP response element-binding protein. The novel HAT inhibitor anacardic acid significantly attenuated dieldrin-induced histone acetylation, Protein kinase C delta proteolytic activation and DNA fragmentation in dopaminergic cells protected against dopaminergic neuronal degeneration in primary mesencephalic neuronal cultures. Furthermore, 30-day exposure of dieldrin in mouse models induced histone hyperacetylation in the striatum and substantia nigra. For the first time, our results collectively demonstrate that exposure to the neurotoxic pesticide dieldrin induces acetylation of core histones because of proteasomal dysfunction and that hyperacetylation plays a key role in dopaminergic neuronal degeneration after exposure of dieldrin.
Collapse
Affiliation(s)
- C Song
- Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | | | | | | | | |
Collapse
|
228
|
Strain-specific BDNF expression of rat primary astrocytes. J Neuroimmunol 2010; 220:90-8. [PMID: 20176405 DOI: 10.1016/j.jneuroim.2010.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Revised: 02/01/2010] [Accepted: 02/02/2010] [Indexed: 12/15/2022]
Abstract
By producing brain-derived neurotrophic factor (BDNF), astrocytes play a role in disease resistance. This study was undertaken to investigate whether primary astrocytes derived from LEW/N and F344/N rats differentially express BDNF. LEW/N astrocytes expressed more BDNF mRNA and protein than F344/N astrocytes in basal and valproic acid (VPA)-stimulated conditions. VPA suppresses HDAC enzyme activity without affecting HDAC gene and protein expression in astrocytes of both strains. Blockade of TrkB receptors resulted in similar fold decreases in basal BDNF mRNA levels between two strains. The results suggest that inhibition of HDAC activity and BDNF-TrkB autocrine loop are involved in regulation of astrocytic BDNF transcription, whereas the mechanisms for elevated constitutive gene BDNF expression of LEW/N astrocytes remain to be investigated.
Collapse
|
229
|
Kim SY, Park E, Park JA, Choi BS, Kim S, Jeong G, Kim CS, Kim DK, Kim SJ, Chun HS. The plant phenolic diterpene carnosol suppresses sodium nitroprusside-induced toxicity in c6 glial cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2010; 58:1543-1550. [PMID: 20073473 DOI: 10.1021/jf903294x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Carnosol, a naturally occurring bioactive phenolic diterpene originating from rosemary and sage, has been shown to exert antioxidant and anti-inflammatory effects. This study examined possible protective effects of carnosol on sodium nitroprusside (SNP)-induced cytotoxicity in C6 glial cells. Carnosol (1-10 microM) dose-dependently attenuated SNP (100 microM)-induced cell death and NO production. SNP-induced apoptotic characteristics, including DNA fragmentation, caspase-3 activation, and c-jun N-terminal protein kinase (JNK) phosphorylation, were significantly suppressed by carnosol (10 microM). In addition, carnosol pretreatment restored the level of reduced glutathione (GSH), which was diminished by SNP treatment. Although both SNP (100 microM) and carnosol (10 microM) stimulated the HO-1 expression time-dependently, SNP caused a temporal increase in HO-1 in early time periods (3-6 h) before cell death occurred. In contrast, carnosol induced the sustained expression of HO-1 until a late time point (24 h). The addition of 1 microM zinc protoporphyrin IX (ZnPP), a specific HO inhibitor, with SNP or carnosol further reduced cell viability. Also, the addition of ZnPP inhibited the protective effect of carnosol against SNP-induced cytotoxicity in C6 cells. These results suggest that carnosol possesses abilities to inhibit SNP-mediated glial cell death through modulation of apoptotic events and induction of HO-1 expression.
Collapse
Affiliation(s)
- Sang Yong Kim
- Department of Biotechnology, Chosun University, Gwangju 501-759, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
230
|
Perisic T, Zimmermann N, Kirmeier T, Asmus M, Tuorto F, Uhr M, Holsboer F, Rein T, Zschocke J. Valproate and amitriptyline exert common and divergent influences on global and gene promoter-specific chromatin modifications in rat primary astrocytes. Neuropsychopharmacology 2010; 35:792-805. [PMID: 19924110 PMCID: PMC3055607 DOI: 10.1038/npp.2009.188] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Revised: 10/22/2009] [Accepted: 10/22/2009] [Indexed: 01/03/2023]
Abstract
Aberrant biochemical processes in the brain frequently go along with subtle shifts of the cellular epigenetic profile that might support the pathogenic progression of psychiatric disorders. Although recent reports have implied the ability of certain antidepressants and mood stabilizers to modulate epigenetic parameters, studies comparing the actions of these compounds under the same conditions are lacking. In this study, we screened amitriptyline (AMI), venlafaxine, citalopram, as well as valproic acid (VPA), carbamazepine, and lamotrigine for their potential actions on global and local epigenetic modifications in rat primary astrocytes. Among all drugs, VPA exposure evoked the strongest global chromatin modifications, including histone H3/H4 hyperacetylation, 2MeH3K9 hypomethylation, and DNA demethylation, as determined by western blot and luminometric methylation analysis, respectively. CpG demethylation occurred independently of DNA methyltransferase (DNMT) suppression. Strikingly, AMI also induced slight cytosine demethylation, paralleled by the reduction in DNMT enzymatic activity, without affecting the global histone acetylation status. Locally, VPA-induced chromatin modifications were reflected at the glutamate transporter (GLT-1) promoter as shown by bisulfite sequencing and acetylated histone H4 chromatin immunoprecipitation analysis. Distinct CpG sites in the distal part of the GLT-1 promoter were demethylated and enriched in acetylated histone H4 in response to VPA. For the first time, we could show that these changes were associated with an enhanced transcription of this astrocyte-specific gene. In contrast, AMI failed to stimulate GLT-1 transcription and to alter promoter methylation levels. In conclusion, VPA and AMI globally exerted chromatin-modulating activities using different mechanisms that divergently precipitated at an astroglial gene locus.
Collapse
Affiliation(s)
- Tatjana Perisic
- Chaperone Research Group, Max-Planck-Institute of Psychiatry, Munich, Bavaria, Germany
| | - Nicole Zimmermann
- Chaperone Research Group, Max-Planck-Institute of Psychiatry, Munich, Bavaria, Germany
| | - Thomas Kirmeier
- Chaperone Research Group, Max-Planck-Institute of Psychiatry, Munich, Bavaria, Germany
| | - Maria Asmus
- Pharmacogenetics Group, Max-Planck-Institute of Psychiatry, Munich, Bavaria, Germany
| | - Francesca Tuorto
- Division of Epigenetics, German Cancer Research Center, Heidelberg, Germany
- Institute of Genetics and Biophysics ‘A. Buzzati-Traverso', CNR, Naples, Italy
| | - Manfred Uhr
- Pharmacogenetics Group, Max-Planck-Institute of Psychiatry, Munich, Bavaria, Germany
| | - Florian Holsboer
- Chaperone Research Group, Max-Planck-Institute of Psychiatry, Munich, Bavaria, Germany
- Pharmacogenetics Group, Max-Planck-Institute of Psychiatry, Munich, Bavaria, Germany
| | - Theo Rein
- Chaperone Research Group, Max-Planck-Institute of Psychiatry, Munich, Bavaria, Germany
| | - Jürgen Zschocke
- Chaperone Research Group, Max-Planck-Institute of Psychiatry, Munich, Bavaria, Germany
| |
Collapse
|
231
|
Dietz KC, Casaccia P. HDAC inhibitors and neurodegeneration: at the edge between protection and damage. Pharmacol Res 2010; 62:11-7. [PMID: 20123018 DOI: 10.1016/j.phrs.2010.01.011] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Accepted: 01/15/2010] [Indexed: 12/22/2022]
Abstract
The use of histone deacetylase inhibitors (HDACIs) as a therapeutic tool for neurodegenerative disorders has been examined with great interest in the last decade. The functional response to treatment with broad-spectrum inhibitors however, has been heterogeneous: protective in some cases and detrimental in others. In this review we discuss potential underlying causes for these apparently contradictory results. Because HDACs are part of repressive complexes, the functional outcome has been characteristically attributed to enhanced gene expression due to increased acetylation of lysine residues on nucleosomal histones. However, it is important to take into consideration that the up-regulation of diverse sets of genes (i.e. pro-apoptotic and anti-apoptotic) may orchestrate different responses in diverse cell types. An alternative possibility is that broad-spectrum pharmacological inhibition may target nuclear or cytosolic HDAC isoforms, with distinct non-histone substrates (i.e. transcription factors; cytoskeletal proteins). Thus, for any given neurological disorder, it is important to take into account the effect of HDACIs on neuronal, glial and inflammatory cells and define the relative contribution of distinct HDAC isoforms to the pathological process. This review article addresses how opposing effects on distinct cell types may profoundly influence the overall therapeutic potential of HDAC inhibitors when investigating treatments for neurodegenerative disorders.
Collapse
Affiliation(s)
- Karen C Dietz
- Department of Neuroscience and Genetics & Genomics, Mount Sinai School of Medicine, One Gustave Levy Place, Box 1065, New York, NY 10029, United States
| | | |
Collapse
|
232
|
Targeting Glial Cells to Elucidate the Pathogenesis of Huntington’s Disease. Mol Neurobiol 2010; 41:248-55. [DOI: 10.1007/s12035-009-8097-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Accepted: 12/27/2009] [Indexed: 10/19/2022]
|
233
|
Neurocognition in bipolar disorders—A closer look at comorbidities and medications. Eur J Pharmacol 2010; 626:87-96. [PMID: 19836378 DOI: 10.1016/j.ejphar.2009.10.018] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2009] [Accepted: 10/09/2009] [Indexed: 01/01/2023]
|
234
|
Abstract
Epigenetics is a rapidly growing field and holds great promise for a range of human diseases, including brain disorders such as Rett syndrome, anxiety and depressive disorders, schizophrenia, Alzheimer disease and Huntington disease. This review is concerned with the pharmacology of epigenetics to treat disorders of the epigenome whether induced developmentally or manifested/acquired later in life. In particular, we will focus on brain disorders and their treatment by drugs that modify the epigenome. While the use of DNA methyl transferase inhibitors and histone deacetylase inhibitors in in vitro and in vivo models have demonstrated improvements in disease-related deficits, clinical trials in humans have been less promising. We will address recent advances in our understanding of the complexity of the epigenome with its many molecular players, and discuss evidence for a compromised epigenome in the context of an ageing or diseased brain. We will also draw on examples of species differences that may exist between humans and model systems, emphasizing the need for more robust pre-clinical testing. Finally, we will discuss fundamental issues to be considered in study design when targeting the epigenome.
Collapse
Affiliation(s)
- Pritika Narayan
- Department of Pharmacology and the National Research Centre for Growth and Development, The University of Auckland, Auckland, New Zealand
| | | |
Collapse
|
235
|
Hung SY, Liou HC, Fu WM. The mechanism of heme oxygenase-1 action involved in the enhancement of neurotrophic factor expression. Neuropharmacology 2009; 58:321-9. [PMID: 19925812 DOI: 10.1016/j.neuropharm.2009.11.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Revised: 11/07/2009] [Accepted: 11/10/2009] [Indexed: 10/20/2022]
Abstract
Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron and bilirubin. Bilirubin is a potent antioxidant and neuroprotectant. Neurotrophic factors of BDNF and GDNF also play important roles in survival and morphological differentiation of dopaminergic neurons. We have previously found that HO-1 induction by adenovirus containing human HO-1 gene (Ad-HO-1) in substantia nigra of rat increases BDNF and GDNF expression. We here further examined the possible mechanism of HO-1 action involved in the enhancement of neurotrophic factor expression. Treatment of anti-BDNF/GDNF antibody significantly enhanced dopaminergic neuronal death, whereas Ad-HO-1 co-treatment was able to antagonize the apoptosis-inducing effect of these antibodies. The confocal imaging shows that HO-1 induction appeared in dopaminergic neuron, astrocyte and microglia at 24 h after injecting Ad-HO-1. HO-1 induced-BDNF/GDNF mRNA expression in substantia nigra was 26/21 folds of that of the contralateral Ad-injected side. The downstream product bilirubin increased GDNF expression through ERK and PI3K-Akt pathways, and also enhanced NFkappaB (p65 and p50) nuclear translocation in glia-enriched cultures. In addition, bilirubin also enhanced BDNF expression through similar pathway in cortical neuron-enriched cultures. We also examined the effect of another HO-1 product, CO, by using CO donor. [Ru(CO)3Cl2]2 increased neurotrophic factor expression via sGC-PKG pathway in both neuron and glia. These results indicate that the downstream products of HO-1, i.e. bilirubin and CO, modulate BDNF and GDNF expression in neuron and astrocyte.
Collapse
Affiliation(s)
- Shih-Ya Hung
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | | | | |
Collapse
|
236
|
Amygdala volume in depressed patients with bipolar disorder assessed using high resolution 3T MRI: the impact of medication. Neuroimage 2009; 49:2966-76. [PMID: 19931399 DOI: 10.1016/j.neuroimage.2009.11.025] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Revised: 11/06/2009] [Accepted: 11/11/2009] [Indexed: 12/24/2022] Open
Abstract
MRI-based reports of both abnormally increased and decreased amygdala volume in bipolar disorder (BD) have surfaced in the literature. Two major methodological weaknesses characterizing extant studies are treatment with medication and inaccurate segmentation of the amygdala due to limitations in spatial and tissue contrast resolution. Here, we acquired high-resolution images (voxel size=0.55 x 0.55 x 0.60 mm) using a GE 3T MRI scanner, and a pulse sequence optimized for tissue contrast resolution. The amygdala was manually segmented by one rater blind to diagnosis, using coronal images. Eighteen unmedicated (mean medication-free period 11+/-10 months) BD subjects were age and gender matched with 18 healthy controls, and 17 medicated (lithium or divalproex) subjects were matched to 17 different controls. The unmedicated BD patients displayed smaller left and right amygdala volumes than their matched control group (p<0.01). Conversely, the BD subjects undergoing medication treatment showed a trend towards greater amygdala volumes than their matched HC sample (p=0.051). Right and left amygdala volumes were larger (p<0.05) or trended larger, respectively, in the medicated BD sample compared with the unmedicated BD sample. The two control groups did not differ from each other in either left or right amygdala volume. BD patients treated with lithium have displayed increased gray matter volume of the cortex and hippocampus relative to untreated BD subjects in previous studies. Here we extend these results to the amygdala. We raise the possibility that neuroplastic changes in the amygdala associated with BD are moderated by some mood stabilizing medications.
Collapse
|
237
|
Hunsberger J, Austin DR, Henter ID, Chen G. The neurotrophic and neuroprotective effects of psychotropic agents. DIALOGUES IN CLINICAL NEUROSCIENCE 2009. [PMID: 19877500 PMCID: PMC2804881 DOI: 10.31887/dcns.2009.11.3/jhunsberger] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Accumulating evidence suggests that psychotropic agents such as mood stabilizers, antidepressants, and antipsychotics realize their neurotrophic/neuroprotective effects by activating the mitogen activated protein kinaselextracellular signal-related kinase, PI3-kinase, and winglesslglycogen synthase kinase (GSK) 3 signaling pathways. These agents also upregulate the expression of trophic/protective molecules such as brain-derived neurotrophic factor, nerve growth factor, B-cell lymphoma 2, serine-threonine kinase, and Bcl-2 associated athanogene 1, and inactivate proapoptotic molecules such as GSK-3, They also promote neurogenesis and are protective in models of neurodegenerative diseases and ischemia. Most if not all, of this evidence was collected from animal studies that used clinically relevant treatment regimens. Furthermore, human imaging studies have found that these agents increase the volume and density of brain tissue, as well as levels of N-acetyl aspartate and glutamate in selected brain regions. Taken together, these data suggest that the neurotrophic/neuroprotective effects of these agents have broad therapeutic potential in the treatment, not only of mood disorders and schizophrenia, but also neurodegenerative diseases and ischemia.
Collapse
Affiliation(s)
- Joshua Hunsberger
- Laboratory of Molecular Pathophysiology and Experimental Therapeutics, Mood and Anxiety Disorders Program, NIMH, NIH, Bethesda, MD, 20892, USA
| | | | | | | |
Collapse
|
238
|
Qian Y, Zheng Y, Tiffany-Castiglioni E. Valproate reversibly reduces neurite outgrowth by human SY5Y neuroblastoma cells. Brain Res 2009; 1302:21-33. [DOI: 10.1016/j.brainres.2009.09.051] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2009] [Revised: 09/10/2009] [Accepted: 09/14/2009] [Indexed: 11/26/2022]
|
239
|
Long-Smith CM, Sullivan AM, Nolan YM. The influence of microglia on the pathogenesis of Parkinson's disease. Prog Neurobiol 2009; 89:277-87. [DOI: 10.1016/j.pneurobio.2009.08.001] [Citation(s) in RCA: 207] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Revised: 08/08/2009] [Accepted: 08/10/2009] [Indexed: 01/03/2023]
|
240
|
Chuang DM, Leng Y, Marinova Z, Kim HJ, Chiu CT. Multiple roles of HDAC inhibition in neurodegenerative conditions. Trends Neurosci 2009; 32:591-601. [PMID: 19775759 PMCID: PMC2771446 DOI: 10.1016/j.tins.2009.06.002] [Citation(s) in RCA: 505] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2009] [Revised: 06/26/2009] [Accepted: 06/26/2009] [Indexed: 12/24/2022]
Abstract
Histone deacetylases (HDACs) play a key role in homeostasis of protein acetylation in histones and other proteins and in regulating fundamental cellular activities such as transcription. A wide range of brain disorders are associated with imbalances in protein acetylation levels and transcriptional dysfunctions. Treatment with various HDAC inhibitors can correct these deficiencies and has emerged as a promising new strategy for therapeutic intervention in neurodegenerative disease. Here, we review and discuss intriguing recent developments in the use of HDAC inhibitors to combat neurodegenerative conditions in cellular and disease models. HDAC inhibitors have neuroprotective, neurotrophic and anti-inflammatory properties; improvements in neurological performance, learning/memory and other disease phenotypes are frequently seen in these models. We discuss the targets and mechanisms underlying these effects of HDAC inhibition and comment on the potential for some HDAC inhibitors to prove clinically effective in the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- De-Maw Chuang
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health, 10 Center Drive MSC 1363, Bethesda, MD 20892-1363, USA.
| | | | | | | | | |
Collapse
|
241
|
Huang HY, Lin SZ, Chen WF, Li KW, Kuo JS, Wang MJ. Urocortin modulates dopaminergic neuronal survival via inhibition of glycogen synthase kinase-3β and histone deacetylase. Neurobiol Aging 2009; 32:1662-77. [PMID: 19875195 DOI: 10.1016/j.neurobiolaging.2009.09.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Revised: 08/19/2009] [Accepted: 09/27/2009] [Indexed: 11/27/2022]
Abstract
Urocortin (UCN) is a member of the corticotropin-releasing hormone (CRH) family of neuropeptides that regulates stress responses. Although UCN is principally expressed in dopaminergic neurons in rat substantia nigra (SN), the function of UCN in modulating dopaminergic neuronal survival remains unclear. Using primary mesencephalic cultures, we demonstrated that dopaminergic neurons underwent spontaneous cell death when their age increased in culture. Treatment of mesencephalic cultures with UCN markedly prolonged the survival of dopaminergic neurons, whereas neutralization of UCN with anti-UCN antibody accelerated dopaminergic neurons degeneration. UCN increased intracellular cAMP levels followed by phosphorylating glycogen synthase kinase-3β (GSK-3β) on Ser9. Moreover, UCN directly inhibited the histone deacetylase (HDAC) activity and induced a robust increase in histone H3 acetylation levels. Using pharmacological approaches, we further demonstrated that inhibition of GSK-3β and HDAC contributes to UCN-mediated neuroprotection. These results suggest that dopaminergic neuron-derived UCN might be involved in an autocrine protective signaling mechanism.
Collapse
Affiliation(s)
- Hsin-Yi Huang
- Department of Research, Neuro-Medical Scientific Center, Buddhist Tzu Chi General Hospital, Hualien 970, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|
242
|
Wang Y, Wang X, Liu L, Wang X. HDAC inhibitor trichostatin A-inhibited survival of dopaminergic neuronal cells. Neurosci Lett 2009; 467:212-6. [PMID: 19835929 DOI: 10.1016/j.neulet.2009.10.037] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Revised: 09/30/2009] [Accepted: 10/09/2009] [Indexed: 11/28/2022]
Abstract
Histone deacetylase (HDAC) inhibitors have been shown associated with neurodegenerative diseases. However, their effects on survival of dopaminergic neurons remain uncertain. In the present study, the HDAC inhibitor trichostatin A (TSA) was tested in following dopaminergic neuronal cell lines: rat N27, mouse MN9D, and human SH-SY5Y cells. Results demonstrated that a single TSA treatment resulted in decreased cell survival and increased apoptosis in dopaminergic neuronal cells. Pre-treatment with TSA resulted in exacerbated neurotoxic damage to dopaminergic neurons induced by 1-methyl-4-phenylpyridinium and rotenone. These results suggest that HDAC inhibitors may influence Parkinson's disease pathogenesis by inhibiting survival and increasing vulnerability of dopaminergic neurons to neurotoxins. Our data also suggested the importance of prudent use of HDAC inhibitors in therapy.
Collapse
Affiliation(s)
- Yong Wang
- Department of Physiology and Key Laboratory of the Neurodegenerative Disorders of the Chinese Ministry of Education, Capital Medical University, Youanmen, Beijing 100069, China
| | | | | | | |
Collapse
|
243
|
Wu HM, Tzeng NS, Qian L, Wei SJ, Hu X, Chen SH, Rawls SM, Flood P, Hong JS, Lu RB. Novel neuroprotective mechanisms of memantine: increase in neurotrophic factor release from astroglia and anti-inflammation by preventing microglial activation. Neuropsychopharmacology 2009; 34:2344-57. [PMID: 19536110 PMCID: PMC3655438 DOI: 10.1038/npp.2009.64] [Citation(s) in RCA: 137] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Memantine shows clinically relevant efficacy in patients with Alzheimer's disease and Parkinson's disease. Most in vivo and in vitro studies attribute the neuroprotective effects of memantine to the blockade of N-methyl-D-aspartate (NMDA) receptor on neurons. However, it cannot be excluded that mechanisms other than NMDA receptor blockade may contribute to the neuroprotective effects of this compound. To address this question, primary midbrain neuron-glia cultures and reconstituted cultures were used, and lipopolysaccharide (LPS), an endotoxin from bacteria, was used to produce inflammation-mediated dopaminergic (DA) neuronal death. Here, we show that memantine exerted both potent neurotrophic and neuroprotective effects on DA neurons in rat neuron-glia cultures. The neurotrophic effect of memantine was glia dependent, as memantine failed to show any positive effect on DA neurons in neuron-enriched cultures. More specifically, it seems to be that astroglia, not microglia, are the source of the memantine-elicited neurotrophic effects through the increased production of glial cell line-derived neurotrophic factor (GDNF). Mechanistic studies showed that GDNF upregulation was associated with histone hyperacetylation by inhibiting the cellular histone deacetylase activity. In addition, memantine also displays neuroprotective effects against LPS-induced DA neuronal damage through its inhibition of microglia activation showed by both OX-42 immunostaining and reduction of pro-inflammatory factor production, such as extracellular superoxide anion, intracellular reactive oxygen species, nitric oxide, prostaglandin E(2), and tumor necrosis factor-alpha. These results suggest that the neuroprotective effects of memantine shown in our cell culture studies are mediated in part through alternative novel mechanisms by reducing microglia-associated inflammation and by stimulating neurotrophic factor release from astroglia.
Collapse
Affiliation(s)
- Hung-Ming Wu
- College of Medicine, Institute of Behavioral Medicine, National Cheng-Kung University, Tainan, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
244
|
Monti B, Gatta V, Piretti F, Raffaelli SS, Virgili M, Contestabile A. Valproic acid is neuroprotective in the rotenone rat model of Parkinson's disease: involvement of alpha-synuclein. Neurotox Res 2009; 17:130-41. [PMID: 19626387 DOI: 10.1007/s12640-009-9090-5] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Revised: 06/25/2009] [Accepted: 07/08/2009] [Indexed: 12/21/2022]
Abstract
Valproic acid (VPA), an established antiepileptic and antimanic drug, has recently emerged as a promising neuroprotective agent. Among its many cellular targets, VPA has been recently demonstrated to be an effective inhibitor of histone deacetylases. Accordingly, we have adopted a schedule of dietary administration (2% VPA added to the chow) that results in a significant inhibition of histone deacetylase activity and in an increase of histone H3 acetylation in brain tissues of 4 weeks-treated rats. We have tested this schedule of VPA treatment in an animal model of Parkinson's disease (PD), in which degeneration of nigro-striatal dopaminergic neurons is obtained through sub-chronic administration of the mitochondrial toxin, rotenone, via osmotic mini pumps implanted to rats. The decrease of the dopaminergic marker tyrosine hydroxylase in substantia nigra and striatum caused by 7 days toxin administration was prevented in VPA-fed rats. VPA treatment also significantly counteracted the death of nigral neurons and the 50% drop of striatal dopamine levels caused by rotenone administration. The PD-marker protein alpha-synuclein decreased, in its native form, in substantia nigra and striatum of rotenone-treated rats, while monoubiquitinated alpha-synuclein increased in the same regions. VPA treatment counteracted both these alpha-synuclein alterations. Furthermore, monoubiquitinated alpha-synuclein increased its localization in nuclei isolated from substantia nigra of rotenone-treated rats, an effect also prevented by VPA treatment. Nuclear localization of alpha-synuclein has been recently described in some models of PD and its neurodegenerative effect has been ascribed to histone acetylation inhibition. Thus, the ability of VPA to increase histone acetylation is a novel candidate mechanism for its neuroprotective action.
Collapse
Affiliation(s)
- Barbara Monti
- Department of Biology, University of Bologna, Bologna, Italy
| | | | | | | | | | | |
Collapse
|
245
|
Lu DY, Yu WH, Yeh WL, Tang CH, Leung YM, Wong KL, Chen YF, Lai CH, Fu WM. Hypoxia-induced matrix metalloproteinase-13 expression in astrocytes enhances permeability of brain endothelial cells. J Cell Physiol 2009; 220:163-73. [PMID: 19241444 DOI: 10.1002/jcp.21746] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
246
|
Sleiman SF, Basso M, Mahishi L, Kozikowski AP, Donohoe ME, Langley B, Ratan RR. Putting the 'HAT' back on survival signalling: the promises and challenges of HDAC inhibition in the treatment of neurological conditions. Expert Opin Investig Drugs 2009; 18:573-84. [PMID: 19388875 PMCID: PMC2731419 DOI: 10.1517/13543780902810345] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Decreased histone acetyltransferase activity and transcriptional dysfunction have been implicated in almost all neurodegenerative conditions. Increasing net histone acetyltransferase activity through inhibition of the histone deacetylases (HDACs) has been shown to be an effective strategy to delay or halt progression of neurological disease in cellular and rodent models. These findings have provided firm rationale for Phase I and Phase II clinical trials of HDAC inhibitors in Huntington's disease, spinal muscular atrophy, and Freidreich's ataxia. In this review, we discuss the current findings and promise of HDAC inhibition as a strategy for treating neurological disorders. Despite the fact that HDAC inhibitors are in an advanced stage of development, we suggest other approaches to modulating HDAC function that may be less toxic and more efficacious than the canonical agents developed so far.
Collapse
Affiliation(s)
- Sama F Sleiman
- Burke Medical Research Institute, 785 Mamaroneck Avenue, White Plains, 10605 NY, USA, Tel: +1 914 368 3121; Fax: +1 914 368 3150
| | - Manuela Basso
- Burke Medical Research Institute, 785 Mamaroneck Avenue, White Plains, 10605 NY, USA, Tel: +1 914 368 3121; Fax: +1 914 368 3150
| | - Lata Mahishi
- Burke Medical Research Institute, 785 Mamaroneck Avenue, White Plains, 10605 NY, USA, Tel: +1 914 368 3121; Fax: +1 914 368 3150
| | - Alan P Kozikowski
- University of Illinois at Chicago, Department of Medicinal Chemistry and Pharmacognosy, 833 S. Wood Street, Chicago, 60612 IL, USA
| | - Mary E Donohoe
- Burke Medical Research Institute, 785 Mamaroneck Avenue, White Plains, 10605 NY, USA, Tel: +1 914 368 3121; Fax: +1 914 368 3150
| | - Brett Langley
- Burke Medical Research Institute, 785 Mamaroneck Avenue, White Plains, 10605 NY, USA, Tel: +1 914 368 3121; Fax: +1 914 368 3150
| | - Rajiv R Ratan
- Burke Medical Research Institute, 785 Mamaroneck Avenue, White Plains, 10605 NY, USA, Tel: +1 914 368 3121; Fax: +1 914 368 3150
| |
Collapse
|
247
|
Lencz T, Lipsky RH, DeRosse P, Burdick KE, Kane JM, Malhotra AK. Molecular differentiation of schizoaffective disorder from schizophrenia using BDNF haplotypes. Br J Psychiatry 2009; 194:313-8. [PMID: 19336781 PMCID: PMC2664969 DOI: 10.1192/bjp.bp.108.050401] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Revised: 05/21/2008] [Accepted: 09/01/2008] [Indexed: 12/14/2022]
Abstract
BACKGROUND Allelic variation in the gene encoding brain-derived neurotrophic factor (BDNF) has been associated with affective disorders, but generally not schizophrenia. Brain-derived neurotrophic factor variants may help clarify the status of schizoaffective disorder. AIMS To test the hypothesis that BDNF haplotypes are associated with psychiatric illness marked by a prominent affective component. METHOD Frequencies of a 5-marker BDNF haplotype were examined in 600 White participants across four diagnostic categories and healthy controls. RESULTS Individuals with schizoaffective disorder and other affective disorders were significantly more likely to carry two copies of the most common BDNF haplotype (containing the valine allele of the Val66Met polymorphism) compared with healthy volunteers. Moreover, when compared with people with schizophrenia, individuals with schizoaffective disorder were significantly more likely to carry two copies of the common haplotype. CONCLUSIONS To our knowledge, this is the first candidate gene study to demonstrate association with schizoaffective disorder but not schizophrenia. Variation in the BDNF gene may be associated with the clinical phenotype of affective dysregulation across several DSM-IV diagnostic categories.
Collapse
Affiliation(s)
- Todd Lencz
- The Zucker Hillside Hospital, Psychiatry Research, 75-59 263rd Street, Glen Oaks, New York 11004, USA.
| | | | | | | | | | | |
Collapse
|
248
|
Creson TK, Yuan P, Manji HK, Chen G. Evidence for involvement of ERK, PI3K, and RSK in induction of Bcl-2 by valproate. J Mol Neurosci 2009; 37:123-34. [PMID: 18677583 PMCID: PMC2788987 DOI: 10.1007/s12031-008-9122-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Accepted: 06/04/2008] [Indexed: 12/01/2022]
Abstract
Valproate, an anticonvulsant and mood stabilizer, up-regulates Bcl-2, a neurotrophic/neuroprotective protein. In this study, we investigated the molecular mechanism through which Bcl-2 is up-regulated by valproate using cultured human neuron-like cells. Valproate, within therapeutically relevant ranges, induced time- and concentration-dependent up-regulations of both Bcl-2 messenger RNA and protein implicating an underlying gene transcriptional-mediated mechanism. Bcl-2 up-regulations were associated with ERK1/2 and PI3K pathway activations and elevated levels of activated phospho-RSK and phospho-CREB, convergent targets of the ERK1/2 and PI3K pathways. Valproate increased transcriptional activity of a human bcl-2 promoter-reporter gene construct. This effect was attenuated, but not blocked, by mutation of a CREB DNA binding site, a CRE site in the human bcl-2 promoter sequence. ERK and/or PI3K pathway inhibitors and RSK1 small hairpin RNA knockdown reduced, but did not abolish, baseline and valproate-induced promoter activities and lowered Bcl-2 protein levels. These data collectively suggest that valproate induces Bcl-2 regulation partially through activations of the ERK and PI3K cascades and their convergent kinase, RSK, although other unknown mechanism(s) are likely involved. Given the known roles of Bcl-2 in the central nervous system, the current findings offer a partial yet complex molecular mechanistic explanation for the known neurobiological effects of valproate including neurite growth, neuronal survival, and neurogenesis.
Collapse
Affiliation(s)
- Thomas K. Creson
- Laboratory of Molecular Pathophysiology, National Institute of Mental Health, National Institutes of Health, 35 Convent Drive, Bldg 35, Rm 1C-912, Bethesda, MD 20892-3711, USA
| | - Peixiong Yuan
- Laboratory of Molecular Pathophysiology, National Institute of Mental Health, National Institutes of Health, 35 Convent Drive, Bldg 35, Rm 1C-912, Bethesda, MD 20892-3711, USA
| | - Husseini K. Manji
- Laboratory of Molecular Pathophysiology, National Institute of Mental Health, National Institutes of Health, 35 Convent Drive, Bldg 35, Rm 1C-912, Bethesda, MD 20892-3711, USA
| | - Guang Chen
- Laboratory of Molecular Pathophysiology, National Institute of Mental Health, National Institutes of Health, 35 Convent Drive, Bldg 35, Rm 1C-912, Bethesda, MD 20892-3711, USA
| |
Collapse
|
249
|
Frisch C, Hüsch K, Angenstein F, Kudin A, Kunz W, Elger CE, Helmstaedter C. Dose-dependent memory effects and cerebral volume changes after in utero exposure to valproate in the rat. Epilepsia 2009; 50:1432-41. [PMID: 19374658 DOI: 10.1111/j.1528-1167.2008.01943.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PURPOSE Recent clinical studies raised concern of a cognitive teratogenicity of the major antiepileptic drug valproate. To investigate possible cerebral correlates, we established a forced self-application schedule by diluting valproate in the drinking water of pregnant Wistar rats. METHODS After application of medium (MD) and high doses (HDs) with mean daily intakes of about 470 and 720 mg/kg during the entire pregnancy, we analyzed effects on offspring performance in a series of behavioral paradigms as well as brain volumetric changes by magnetic resonance imaging (MRI). RESULTS While high dosages with peak serum concentrations slightly above 100 microg/ml induced early decrements in general activity and deficits in learning and memory, medium dosages led to improved watermaze performance in 30-day-old rats. MRI analyses indicated increased hippocampal volumes in the MD condition, whereas in the HD condition significantly decreased cortical and brainstem volumes were registered. Cortical volume reduction was correlated with spatial acuity in the watermaze. CONCLUSIONS The results indicate that effects of valproate in utero on offspring cognitive capabilities might depend on total drug load differentially affecting cerebral development during adolescence in the rat.
Collapse
|
250
|
Effects of engrafted neural stem cells in Alzheimer's disease rats. Neurosci Lett 2008; 450:167-71. [PMID: 19070649 DOI: 10.1016/j.neulet.2008.12.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2008] [Revised: 12/03/2008] [Accepted: 12/03/2008] [Indexed: 12/12/2022]
Abstract
Cell therapy is thought to have a central role in restorative therapy, which aims to restore the function of the damaged nervous system. Neural stem cells (NSCs) can differentiate into neurons, astrocytes and oligodendrocytes. The purpose of this study was to evaluate the therapeutic effects of transplanting NSCs into rats which have the animal model of Alzheimer's disease (AD). NSCs from the hippocampus and NSCs-derived glial cells labeled with 5'-Bromo-2'-deoxyuridine (BrdU) were transplanted into two groups of transected rat basal forebrain. Nestin staining, glial fibrillary acidic protein (GFAP) staining and double-labeling immunofluorescence were used to detect the engrafted cells in the basal forebrain. Immunohistochemical detection of p75(NGFR) showed that the number of cholinergic neurons of the NSCs-transplanted group was significant higher than that of the glia-transplanted group in medial septum (MS) and vertical diagonal branch (VDB) (P<0.05). Learning and memory abilities were also measured by Y-maze test. The results indicate that transplanted NSCs can differentiate into cholinergic neurons, which may play an important role in the therapeutic effects of transplanted NSCs.
Collapse
|