1
|
Ahtiainen A, Genocchi B, Subramaniyam NP, Tanskanen JMA, Rantamäki T, Hyttinen JAK. Astrocytes facilitate gabazine-evoked electrophysiological hyperactivity and distinct biochemical responses in mature neuronal cultures. J Neurochem 2024; 168:3076-3094. [PMID: 39001671 DOI: 10.1111/jnc.16182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/07/2024] [Accepted: 07/03/2024] [Indexed: 10/04/2024]
Abstract
Gamma-aminobutyric acid (GABA) is the principal inhibitory neurotransmitter in the adult brain that binds to GABA receptors and hyperpolarizes the postsynaptic neuron. Gabazine acts as a competitive antagonist to type A GABA receptors (GABAAR), thereby causing diminished neuronal hyperpolarization and GABAAR-mediated inhibition. However, the biochemical effects and the potential regulatory role of astrocytes in this process remain poorly understood. To address this, we investigated the neuronal responses of gabazine in rat cortical cultures containing varying ratios of neurons and astrocytes. Electrophysiological characterization was performed utilizing microelectrode arrays (MEAs) with topologically controlled microcircuit cultures that enabled control of neuronal network growth. Biochemical analysis of the cultures was performed using traditional dissociated cultures on coverslips. Our study indicates that, upon gabazine stimulation, astrocyte-rich neuronal cultures exhibit elevated electrophysiological activity and tyrosine phosphorylation of tropomyosin receptor kinase B (TrkB; receptor for brain-derived neurotrophic factor), along with distinct cytokine secretion profiles. Notably, neurons lacking proper astrocytic support were found to experience synapse loss and decreased mitogen-activated protein kinase (MAPK) phosphorylation. Furthermore, astrocytes contributed to neuronal viability, morphology, vascular endothelial growth factor (VEGF) secretion, and overall neuronal network functionality, highlighting the multifunctional role of astrocytes.
Collapse
Affiliation(s)
- Annika Ahtiainen
- Computational Biophysics and Imaging Group, BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Barbara Genocchi
- Computational Biophysics and Imaging Group, BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Narayan Puthanmadam Subramaniyam
- Computational Biophysics and Imaging Group, BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jarno M A Tanskanen
- Computational Biophysics and Imaging Group, BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Tomi Rantamäki
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- SleepWell Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jari A K Hyttinen
- Computational Biophysics and Imaging Group, BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
2
|
Modifiable risk factors of dementia linked to excitation-inhibition imbalance. Ageing Res Rev 2023; 83:101804. [PMID: 36410620 DOI: 10.1016/j.arr.2022.101804] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 11/04/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022]
Abstract
Recent evidence identifies 12 potentially modifiable risk factors for dementia to which 40% of dementia cases are attributed. While the recognition of these risk factors has paved the way for the development of new prevention measures, the link between these risk factors and the underlying pathophysiology of dementia is yet not well understood. A growing number of recent clinical and preclinical studies support a role of Excitation-Inhibition (E-I) imbalance in the pathophysiology of dementia. In this review, we aim to propose a conceptual model on the links between the modifiable risk factors and the E-I imbalance in dementia. This model, which aims to address the current gap in the literature, is based on 12 mediating common mechanisms: the hypothalamic-pituitary-adrenal (HPA) axis dysfunction, neuroinflammation, oxidative stress, mitochondrial dysfunction, cerebral hypo-perfusion, blood-brain barrier (BBB) dysfunction, beta-amyloid deposition, elevated homocysteine level, impaired neurogenesis, tau tangles, GABAergic dysfunction, and glutamatergic dysfunction. We believe this model serves as a framework for future studies in this field and facilitates future research on dementia prevention, discovery of new biomarkers, and developing new interventions.
Collapse
|
3
|
Ali AB, Islam A, Constanti A. The fate of interneurons, GABA A receptor sub-types and perineuronal nets in Alzheimer's disease. Brain Pathol 2022; 33:e13129. [PMID: 36409151 PMCID: PMC9836378 DOI: 10.1111/bpa.13129] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/12/2022] [Indexed: 11/23/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurological disease, which is associated with gradual memory loss and correlated with synaptic hyperactivity and abnormal oscillatory rhythmic brain activity that precedes phenotypic alterations and is partly responsible for the spread of the disease pathology. Synaptic hyperactivity is thought to be because of alteration in the homeostasis of phasic and tonic synaptic inhibition, which is orchestrated by the GABAA inhibitory system, encompassing subclasses of interneurons and GABAA receptors, which play a vital role in cognitive functions, including learning and memory. Furthermore, the extracellular matrix, the perineuronal nets (PNNs) which often go unnoticed in considerations of AD pathology, encapsulate the inhibitory cells and neurites in critical brain regions and have recently come under the light for their crucial role in synaptic stabilisation and excitatory-inhibitory balance and when disrupted, serve as a potential trigger for AD-associated synaptic imbalance. Therefore, in this review, we summarise the current understanding of the selective vulnerability of distinct interneuron subtypes, their synaptic and extrasynaptic GABAA R subtypes as well as the changes in PNNs in AD, detailing their contribution to the mechanisms of disease development. We aim to highlight how seemingly unique malfunction in each component of the interneuronal GABA inhibitory system can be tied together to result in critical circuit dysfunction, leading to the irreversible symptomatic damage observed in AD.
Collapse
|
4
|
Key Genes and Biochemical Networks in Various Brain Regions Affected in Alzheimer's Disease. Cells 2022; 11:cells11060987. [PMID: 35326437 PMCID: PMC8946735 DOI: 10.3390/cells11060987] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/02/2022] [Accepted: 03/10/2022] [Indexed: 12/27/2022] Open
Abstract
Alzheimer’s disease (AD) is one of the most complicated progressive neurodegenerative brain disorders, affecting millions of people around the world. Ageing remains one of the strongest risk factors associated with the disease and the increasing trend of the ageing population globally has significantly increased the pressure on healthcare systems worldwide. The pathogenesis of AD is being extensively investigated, yet several unknown key components remain. Therefore, we aimed to extract new knowledge from existing data. Ten gene expression datasets from different brain regions including the hippocampus, cerebellum, entorhinal, frontal and temporal cortices of 820 AD cases and 626 healthy controls were analyzed using the robust rank aggregation (RRA) method. Our results returned 1713 robust differentially expressed genes (DEGs) between five brain regions of AD cases and healthy controls. Subsequent analysis revealed pathways that were altered in each brain region, of which the GABAergic synapse pathway and the retrograde endocannabinoid signaling pathway were shared between all AD affected brain regions except the cerebellum, which is relatively less sensitive to the effects of AD. Furthermore, we obtained common robust DEGs between these two pathways and predicted three miRNAs as potential candidates targeting these genes; hsa-mir-17-5p, hsa-mir-106a-5p and hsa-mir-373-3p. Three transcription factors (TFs) were also identified as the potential upstream regulators of the robust DEGs; ELK-1, GATA1 and GATA2. Our results provide the foundation for further research investigating the role of these pathways in AD pathogenesis, and potential application of these miRNAs and TFs as therapeutic and diagnostic targets.
Collapse
|
5
|
Hammoud H, Netsyk O, Tafreshiha AS, Korol SV, Jin Z, Li J, Birnir B. Insulin differentially modulates GABA signalling in hippocampal neurons and, in an age-dependent manner, normalizes GABA-activated currents in the tg-APPSwe mouse model of Alzheimer's disease. Acta Physiol (Oxf) 2021; 232:e13623. [PMID: 33559388 DOI: 10.1111/apha.13623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/29/2021] [Accepted: 02/03/2021] [Indexed: 02/06/2023]
Abstract
AIM We examined if tonic γ-aminobutyric acid (GABA)-activated currents in primary hippocampal neurons were modulated by insulin in wild-type and tg-APPSwe mice, an Alzheimer's disease (AD) model. METHODS GABA-activated currents were recorded in dentate gyrus (DG) granule cells and CA3 pyramidal neurons in hippocampal brain slices, from 8 to 10 weeks old (young) wild-type mice and in dorsal DG granule cells in adult, 5-6 and 10-12 (aged) months old wild-type and tg-APPSwe mice, in the absence or presence of insulin, by whole-cell patch-clamp electrophysiology. RESULTS In young mice, insulin (1 nmol/L) enhanced the total spontaneous inhibitory postsynaptic current (sIPSCT ) density in both dorsal and ventral DG granule cells. The extrasynaptic current density was only increased by insulin in dorsal CA3 pyramidal neurons. In absence of action potentials, insulin enhanced DG granule cells and dorsal CA3 pyramidal neurons miniature IPSC (mIPSC) frequency, consistent with insulin regulation of presynaptic GABA release. sIPSCT densities in DG granule cells were similar in wild-type and tg-APPSwe mice at 5-6 months but significantly decreased in aged tg-APPSwe mice where insulin normalized currents to wild-type levels. The extrasynaptic current density was increased in tg-APPSwe mice relative to wild-type littermates but, only in aged tg-APPSwe mice did insulin decrease and normalize the current. CONCLUSION Insulin effects on GABA signalling in hippocampal neurons are selective while multifaceted and context-based. Not only is the response to insulin related to cell-type, hippocampal axis-location, age of animals and disease but also to the subtype of neuronal inhibition involved, synaptic or extrasynaptic GABAA receptors-activated currents.
Collapse
Affiliation(s)
- Hayma Hammoud
- Department of Medical Cell Biology Uppsala University Uppsala Sweden
| | - Olga Netsyk
- Department of Medical Cell Biology Uppsala University Uppsala Sweden
| | | | - Sergiy V. Korol
- Department of Medical Cell Biology Uppsala University Uppsala Sweden
| | - Zhe Jin
- Department of Medical Cell Biology Uppsala University Uppsala Sweden
| | - Jin‐Ping Li
- Department of Medical Biochemistry and Microbiology Uppsala University Uppsala Sweden
| | - Bryndis Birnir
- Department of Medical Cell Biology Uppsala University Uppsala Sweden
| |
Collapse
|
6
|
Impaired Expression of GABA Signaling Components in the Alzheimer's Disease Middle Temporal Gyrus. Int J Mol Sci 2020; 21:ijms21228704. [PMID: 33218044 PMCID: PMC7698927 DOI: 10.3390/ijms21228704] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 12/26/2022] Open
Abstract
γ-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter, playing a central role in the regulation of cortical excitability and the maintenance of the excitatory/inhibitory (E/I) balance. Several lines of evidence point to a remodeling of the cerebral GABAergic system in Alzheimer’s disease (AD), with past studies demonstrating alterations in GABA receptor and transporter expression, GABA synthesizing enzyme activity and focal GABA concentrations in post-mortem tissue. AD is a chronic neurodegenerative disorder with a poorly understood etiology and the temporal cortex is one of the earliest regions in the brain to be affected by AD neurodegeneration. Utilizing NanoString nCounter analysis, we demonstrate here the transcriptional downregulation of several GABA signaling components in the post-mortem human middle temporal gyrus (MTG) in AD, including the GABAA receptor α1, α2, α3, α5, β1, β2, β3, δ, γ2, γ3, and θ subunits and the GABAB receptor 2 (GABABR2) subunit. In addition to this, we note the transcriptional upregulation of the betaine-GABA transporter (BGT1) and GABA transporter 2 (GAT2), and the downregulation of the 67 kDa isoform of glutamate decarboxylase (GAD67), the primary GABA synthesizing enzyme. The functional consequences of these changes require further investigation, but such alterations may underlie disruptions to the E/I balance that are believed to contribute to cognitive decline in AD.
Collapse
|
7
|
Barker JS, Hines RM. Regulation of GABA A Receptor Subunit Expression in Substance Use Disorders. Int J Mol Sci 2020; 21:ijms21124445. [PMID: 32580510 PMCID: PMC7352578 DOI: 10.3390/ijms21124445] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 01/02/2023] Open
Abstract
The modulation of neuronal cell firing is mediated by the release of the neurotransmitter GABA (γ-aminobuytric acid), which binds to two major families of receptors. The ionotropic GABAA receptors (GABAARs) are composed of five distinct subunits that vary in expression by brain region and cell type. The action of GABA on GABAARs is modulated by a variety of clinically and pharmacologically important drugs such as benzodiazepines and alcohol. Exposure to and abuse of these substances disrupts homeostasis and induces plasticity in GABAergic neurotransmission, often via the regulation of receptor expression. Here, we review the regulation of GABAAR subunit expression in adaptive and pathological plasticity, with a focus on substance use. We examine the factors influencing the expression of GABAAR subunit genes including the regulation of the 5′ and 3′ untranslated regions, variations in DNA methylation, immediate early genes and transcription factors that regulate subunit expression, translational and post-translational modifications, and other forms of receptor regulation beyond expression. Advancing our understanding of the factors regulating GABAAR subunit expression during adaptive plasticity, as well as during substance use and withdrawal will provide insight into the role of GABAergic signaling in substance use disorders, and contribute to the development of novel targeted therapies.
Collapse
|
8
|
Bi D, Wen L, Wu Z, Shen Y. GABAergic dysfunction in excitatory and inhibitory (E/I) imbalance drives the pathogenesis of Alzheimer's disease. Alzheimers Dement 2020; 16:1312-1329. [PMID: 32543726 DOI: 10.1002/alz.12088] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 01/28/2020] [Accepted: 02/10/2020] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To propose a new hypothesis that GABAergic dysfunction in excitatory and inhibitory (E/I) imbalance drives the pathogenesis of Alzheimer's disease (AD). BACKGROUND Synaptic dysfunction and E/I imbalance emerge decades before the appearance of cognitive decline in AD patients, which contribute to neurodegeneration. Initially, E/I imbalance was thought to occur first, due to dysfunction of the glutamatergic and cholinergic systems. However, new evidence has demonstrated that the GABAergic system, the counterpart of E/I balance and the major inhibitory neurotransmitter system in the central nervous system, is altered enormously and that this contributes to E/I imbalance and further AD pathogenesis. NEW HYPOTHESIS Alterations to the GABAergic system, induced by multiple AD pathogenic or risk factors, contribute to E/I imbalance and AD pathogenesis. MAJOR CHALLENGES FOR THE HYPOTHESIS This GABAergic hypothesis accounts for many critical questions and common challenges confronting a new hypothesis of AD pathogenesis. More specifically, it explains why amyloid beta (Aβ), β-secretase (BACE1), apolipoprotein E4 gene (APOE ε4), hyperactive glia cells, contributes to AD pathogenesis and why age and sex are the risk factors of AD. GABAergic dysfunction promotes the spread of Aβ pathology throughout the AD brain and associated cognitive impairments, and the induction of dysfunction induced by these varied risk factors shares this common neurobiology leading to E/I imbalance. In turn, some of these factors exacerbate GABAergic dysfunction and E/I imbalance. Moreover, the GABAergic system modulates various brain functions and thus, the GABAergic hypothesis accounts for nonamnestic manifestations. Furthermore, corrections of E/I balance through manipulation of GABAergic functions have shown positive outcomes in preclinical and clinical studies, suggesting the potential of the GABAergic system as a therapeutic target in AD. LINKAGE TO OTHER MAJOR THEORIES Dysfunction of the GABAergic system is induced by multiple critical signaling pathways, which include the existing major theories of AD pathogenesis, such as the Aβ and neuroinflammation hypotheses. In a new perspective, this GABAergic hypothesis accounts for the E/I imbalance and related excitotoxicity, which contribute to cognitive decline and AD pathogenesis. Therefore, the GABAergic system could be a key target to restore, at least partially, the E/I balance and cognitive function in AD patients.
Collapse
Affiliation(s)
- Danlei Bi
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Institute on Aging and Brain Disorders, University of Sciences and Technology of China, Hefei, China.,Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lang Wen
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Institute on Aging and Brain Disorders, University of Sciences and Technology of China, Hefei, China.,Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zujun Wu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Institute on Aging and Brain Disorders, University of Sciences and Technology of China, Hefei, China.,Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yong Shen
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Institute on Aging and Brain Disorders, University of Sciences and Technology of China, Hefei, China.,Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
9
|
Bengtsson S, Bäckström T, Brinton R, Irwin R, Johansson M, Sjöstedt J, Wang M. GABA-A receptor modulating steroids in acute and chronic stress; relevance for cognition and dementia? Neurobiol Stress 2020; 12:100206. [PMID: 31921942 PMCID: PMC6948369 DOI: 10.1016/j.ynstr.2019.100206] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/13/2019] [Accepted: 12/18/2019] [Indexed: 01/10/2023] Open
Abstract
Cognitive dysfunction, dementia and Alzheimer's disease (AD) are increasing as the population worldwide ages. Therapeutics for these conditions is an unmet need. This review focuses on the role of the positive GABA-A receptor modulating steroid allopregnanolone (APα), it's role in underlying mechanisms for impaired cognition and of AD, and to determine options for therapy of AD. On one hand, APα given intermittently promotes neurogenesis, decreases AD-related pathology and improves cognition. On the other, continuous exposure of APα impairs cognition and deteriorates AD pathology. The disparity between these two outcomes led our groups to analyze the mechanisms underlying the difference. We conclude that the effects of APα depend on administration pattern and that chronic slightly increased APα exposure is harmful to cognitive function and worsens AD pathology whereas single administrations with longer intervals improve cognition and decrease AD pathology. These collaborative assessments provide insights for the therapeutic development of APα and APα antagonists for AD and provide a model for cross laboratory collaborations aimed at generating translatable data for human clinical trials.
Collapse
Affiliation(s)
- S.K.S. Bengtsson
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, University of Umeå, Sweden
| | - T. Bäckström
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, University of Umeå, Sweden
| | - R. Brinton
- Center for Innovation in Brain Science, Professor Departments of Pharmacology and Neurology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - R.W. Irwin
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089, USA
| | - M. Johansson
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, University of Umeå, Sweden
| | - J. Sjöstedt
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, University of Umeå, Sweden
| | - M.D. Wang
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, University of Umeå, Sweden
| |
Collapse
|
10
|
Kwakowsky A, Calvo-Flores Guzmán B, Pandya M, Turner C, Waldvogel HJ, Faull RL. GABA A receptor subunit expression changes in the human Alzheimer's disease hippocampus, subiculum, entorhinal cortex and superior temporal gyrus. J Neurochem 2019; 145:374-392. [PMID: 29485232 DOI: 10.1111/jnc.14325] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/17/2018] [Accepted: 02/12/2018] [Indexed: 12/14/2022]
Abstract
Gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the central nervous system. GABA type A receptors (GABAA Rs) are severely affected in Alzheimer's disease (AD). However, the distribution and subunit composition of GABAA Rs in the AD brain are not well understood. This is the first comprehensive study to show brain region- and cell layer-specific alterations in the expression of the GABAA R subunits α1-3, α5, β1-3 and γ2 in the human AD hippocampus, entorhinal cortex and superior temporal gyrus. In late-stage AD tissue samples using immunohistochemistry we found significant alteration of all investigated GABAA Rs subunits except for α3 and β1 that were well preserved. The most prominent changes include an increase in GABAA R α1 expression associated with AD in all layers of the CA3 region, in the stratum (str.) granulare and hilus of the dentate gyrus. We found a significant increase in GABAA R α2 expression in the str. oriens of the CA1-3, str. radiatum of the CA2,3 and decrease in the str. pyramidale of the CA1 region in AD cases. In AD there was a significant increase in GABAA R α5 subunit expression in str. pyramidale, str. oriens of the CA1 region and decrease in the superior temporal gyrus. We also found a significant decrease in the GABAA R β3 subunit immunoreactivity in the str. oriens of the CA2, str. granulare and str. moleculare of the dentate gyrus. In conclusion, these findings indicate that the expression of the GABAA R subunits shows brain region- and layer-specific alterations in AD, and these changes could significantly influence and alter GABAA R function in the disease. Cover Image for this issue: doi: 10.1111/jnc.14179.
Collapse
Affiliation(s)
- Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Beatriz Calvo-Flores Guzmán
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Madhavi Pandya
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Clinton Turner
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Department of Anatomical Pathology, LabPlus, Auckland City Hospital, Auckland, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard L Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
11
|
Zhang T, Li J, Yu H, Shi Y, Li Z, Wang L, Wang Z, Lu T, Wang L, Yue W, Zhang D. Meta-analysis of GABRB2 polymorphisms and the risk of schizophrenia combined with GWAS data of the Han Chinese population and psychiatric genomics consortium. PLoS One 2018; 13:e0198690. [PMID: 29894498 PMCID: PMC5997335 DOI: 10.1371/journal.pone.0198690] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 05/23/2018] [Indexed: 12/31/2022] Open
Abstract
Schizophrenia (SCZ) is a severe psychiatric disorder with evidence of a strong genetic component in the complex etiologies. Some studies indicated that gamma-aminobutyric acid (GABA)A receptor β2 subunit gene (GABRB2) was associated with SCZ. Other studies reported a negative association. Moreover, the results of two previous meta-analyses of GABRB2 with SCZ were inconsistent and the sample sizes were limited. Therefore, an updated meta-analysis combined with genome-wide association study (GWAS) data of the Han Chinese population and Psychiatric Genomics Consortium (PGC) was performed. Available case–control and family-based genetic data were extracted from association studies, and the GWAS data were included. The findings showed no association between six single-nucleotide polymorphisms of GABRB2 (rs6556547, rs1816071, rs1816072, rs194072, rs252944, and rs187269) and SCZ in a total of 51,491 patients and 74,667 controls. The ethnic subgroup analysis revealed no significant association in Asian populations. Since the PGC data of SCZ (SCZ-PGC, 2014) contained 3 studies of Asian populations (1866 patients and 3418 controls), only the data of European samples in SCZ-PGC were used for the meta-analysis of the Caucasian population in the present study. The result still showed no association in the Caucasian population. In conclusion, the present meta-analysis on combined data from GWASs of the Han Chinese population and PGC suggested that GABRB2 polymorphisms might not be associated with SCZ.
Collapse
Affiliation(s)
- Tian Zhang
- Peking University Sixth Hospital, Beijing, China
- Peking University Institute of Mental Health, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
- National Clinical Research Center for Mental Disorders, (Peking University Sixth Hospital), Beijing, China
| | - Jun Li
- Peking University Sixth Hospital, Beijing, China
- Peking University Institute of Mental Health, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
- National Clinical Research Center for Mental Disorders, (Peking University Sixth Hospital), Beijing, China
| | - Hao Yu
- Department of Psychiatry, Jining Medical University, Jining, Shandong, China
| | - Yongyong Shi
- Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
- Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
- Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Psychiatry, First Teaching Hospital of Xinjiang Medical University, Urumqi, China
- Changning Mental Health Center, Shanghai, China
| | - Zhiqiang Li
- Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
- Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
- Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Linyan Wang
- Peking University Sixth Hospital, Beijing, China
- Peking University Institute of Mental Health, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
- National Clinical Research Center for Mental Disorders, (Peking University Sixth Hospital), Beijing, China
| | - Ziqi Wang
- Peking University Sixth Hospital, Beijing, China
- Peking University Institute of Mental Health, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
- National Clinical Research Center for Mental Disorders, (Peking University Sixth Hospital), Beijing, China
| | - Tianlan Lu
- Peking University Sixth Hospital, Beijing, China
- Peking University Institute of Mental Health, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
- National Clinical Research Center for Mental Disorders, (Peking University Sixth Hospital), Beijing, China
| | - Lifang Wang
- Peking University Sixth Hospital, Beijing, China
- Peking University Institute of Mental Health, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
- National Clinical Research Center for Mental Disorders, (Peking University Sixth Hospital), Beijing, China
- * E-mail: (LW); (WY); (DZ)
| | - Weihua Yue
- Peking University Sixth Hospital, Beijing, China
- Peking University Institute of Mental Health, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
- National Clinical Research Center for Mental Disorders, (Peking University Sixth Hospital), Beijing, China
- * E-mail: (LW); (WY); (DZ)
| | - Dai Zhang
- Peking University Sixth Hospital, Beijing, China
- Peking University Institute of Mental Health, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
- National Clinical Research Center for Mental Disorders, (Peking University Sixth Hospital), Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
- * E-mail: (LW); (WY); (DZ)
| |
Collapse
|
12
|
Reyes-Marin KE, Nuñez A. Seizure susceptibility in the APP/PS1 mouse model of Alzheimer's disease and relationship with amyloid β plaques. Brain Res 2017; 1677:93-100. [PMID: 28963050 DOI: 10.1016/j.brainres.2017.09.026] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 09/21/2017] [Accepted: 09/22/2017] [Indexed: 11/17/2022]
Abstract
Alzheimer's disease is a common age associated neurodegenerative disorder associated with an elevated risk of seizures that may be fundamentally connected to cognitive dysfunction. We used 4-9month-old mice of the APP/PS1 mouse model of Alzheimer's disease to study the presence of epileptiform-like discharges and to establish if the amyloid-β plaques affect their generation. The EEG of the APP/PS1 transgenic mice revealed a higher incidence of epileptiform-like discharges i.e. seizure events (interictal spikes, sharp waves, or polyspikes) than in the controls. Also, APP/PS1 mice showed a lower latency to evoke seizure events than in the control animals when pentylenetetrazole (60mg/kg; i.p.) was injected. Moreover, physostigmine injection (1mg/kg; i.p.) also increased the frequency of spontaneous epileptiform-like discharges in the APP/PS1 mice. We also found a correlation between the frequency of epileptiform-like discharges and the number of amyloid-β plaques. Application of N-(2-chloroethyl)-N-ethyl-bromobenzylamine (50mg/kg) generated amyloid-β plaques in the cortex and seizure activity appeared. Taken together, these data indicate that deposits of amyloid-β plaques may be responsible for the epileptiform-like discharges recorded in the APP/PS1 mice and could be responsible for the elevated risk for seizures of Alzheimer's patients.
Collapse
Affiliation(s)
- Karen E Reyes-Marin
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Angel Nuñez
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
13
|
Towards a Better Understanding of GABAergic Remodeling in Alzheimer's Disease. Int J Mol Sci 2017; 18:ijms18081813. [PMID: 28825683 PMCID: PMC5578199 DOI: 10.3390/ijms18081813] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 08/16/2017] [Accepted: 08/17/2017] [Indexed: 12/18/2022] Open
Abstract
γ-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the vertebrate brain. In the past, there has been a major research drive focused on the dysfunction of the glutamatergic and cholinergic neurotransmitter systems in Alzheimer’s disease (AD). However, there is now growing evidence in support of a GABAergic contribution to the pathogenesis of this neurodegenerative disease. Previous studies paint a complex, convoluted and often inconsistent picture of AD-associated GABAergic remodeling. Given the importance of the GABAergic system in neuronal function and homeostasis, in the maintenance of the excitatory/inhibitory balance, and in the processes of learning and memory, such changes in GABAergic function could be an important factor in both early and later stages of AD pathogenesis. Given the limited scope of currently available therapies in modifying the course of the disease, a better understanding of GABAergic remodeling in AD could open up innovative and novel therapeutic opportunities.
Collapse
|
14
|
Li Y, Sun H, Chen Z, Xu H, Bu G, Zheng H. Implications of GABAergic Neurotransmission in Alzheimer's Disease. Front Aging Neurosci 2016; 8:31. [PMID: 26941642 PMCID: PMC4763334 DOI: 10.3389/fnagi.2016.00031] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 02/08/2016] [Indexed: 01/02/2023] Open
Abstract
Alzheimer's disease (AD) is characterized pathologically by the deposition of β-amyloid peptides (Aβ) and the accumulation of neurofibrillary tangles (NFTs) composed of hyper-phosphorylated tau. Regardless of the pathological hallmarks, synaptic dysfunction is widely accepted as a causal event in AD. Of the two major types of synapses in the central nervous system (CNS): glutamatergic and GABAergic, which provide excitatory and inhibitory outputs respectively, abundant data implicate an impaired glutamatergic system during disease progression. However, emerging evidence supports the notion that disrupted default neuronal network underlies impaired memory, and that alterations of GABAergic circuits, either plays a primary role or as a compensatory response to excitotoxicity, may also contribute to AD by disrupting the overall network function. The goal of this review is to provide an overview of the involvement of Aβ, tau and apolipoprotein E4 (apoE4), the major genetic risk factor in late-onset AD (LOAD), in GABAergic neurotransmission and the potential of modulating the GABAergic function as AD therapy.
Collapse
Affiliation(s)
- Yanfang Li
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University Xiamen, China
| | - Hao Sun
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University Xiamen, China
| | - Zhicai Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University Xiamen, China
| | - Huaxi Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen UniversityXiamen, China; Neurodegenerative Disease Research Program, Sanford-Burnham Medical Research InstituteLa Jolla, CA, USA
| | - Guojun Bu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen UniversityXiamen, China; Department of Neuroscience, Mayo ClinicJacksonville, FL, USA
| | - Hui Zheng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen UniversityXiamen, China; The Interdepartmental Program of Translational Biology and Molecular Medicine, Huffington Center on Aging, Baylor College of MedicineHouston, TX, USA
| |
Collapse
|
15
|
Shao H, Zhang Y, Dong Y, Yu B, Xia W, Xie Z. Chronic treatment with anesthetic propofol improves cognitive function and attenuates caspase activation in both aged and Alzheimer's disease transgenic mice. J Alzheimers Dis 2015; 41:499-513. [PMID: 24643139 DOI: 10.3233/jad-132792] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
There is a need to seek new treatment(s) for Alzheimer's disease (AD). A recent study showed that AD patients may have decreased levels of functional GABA receptors. Propofol, a commonly used anesthetic, is a GABA receptor agonist. We therefore set out to perform a proof of concept study to determine whether chronic treatment with propofol (50 mg/kg/week) can improve cognitive function in both aged wild-type (WT) and AD transgenic (Tg) mice. Propofol was administrated to the WT and AD Tg mice once a week for 8 or 12 weeks, respectively. Morris water maze was used to assess the cognitive function of the mice following the propofol treatment. Activation of caspase-3, caspase-9, and caspase-8 was investigated using western blot analysis at the end of the propofol treatment. In the mechanistic studies, effects of propofol, amyloid-β protein (Aβ), and GABA receptor antagonist flumazenil on caspase-3 activation and opening of the mitochondrial permeability transition pore were assessed in H4 human neuroglioma and mouse neuroblastoma cells by western blot analysis and flow cytometry. Here we showed that the propofol treatment improved cognitive function and attenuated brain caspase-3 and caspase-9 activation in both aged WT and AD Tg mice. Propofol attenuated Aβ-induced caspase-3 activation and opening of the mitochondrial permeability transition pore in the cells, and flumazenil inhibited the propofol's effects. These results suggested that propofol might improve cognitive function via attenuating the Aβ-induced mitochondria dysfunction and caspase activation, which explored the potential that anesthetic propofol could improve cognitive function in elderly and AD patients.
Collapse
Affiliation(s)
- Haijun Shao
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.,Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yiying Zhang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Yuanlin Dong
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Buwei Yu
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Weiming Xia
- Department of Veterans Affairs, Medical Research and Development Service and Geriatric Research, Education and Clinical Center, Bedford, MA, USA
| | - Zhongcong Xie
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
16
|
Dihydromyricetin Ameliorates Behavioral Deficits and Reverses Neuropathology of Transgenic Mouse Models of Alzheimer’s Disease. Neurochem Res 2014; 39:1171-81. [DOI: 10.1007/s11064-014-1304-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 04/01/2014] [Accepted: 04/03/2014] [Indexed: 11/27/2022]
|
17
|
Loss of functional GABA(A) receptors in the Alzheimer diseased brain. Proc Natl Acad Sci U S A 2012; 109:10071-6. [PMID: 22691495 DOI: 10.1073/pnas.1204606109] [Citation(s) in RCA: 205] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The cholinergic and glutamatergic neurotransmission systems are known to be severely disrupted in Alzheimer's disease (AD). GABAergic neurotransmission, in contrast, is generally thought to be well preserved. Evidence from animal models and human postmortem tissue suggest GABAergic remodeling in the AD brain. Nevertheless, there is no information on changes, if any, in the electrophysiological properties of human native GABA receptors as a consequence of AD. To gain such information, we have microtransplanted cell membranes, isolated from temporal cortices of control and AD brains, into Xenopus oocytes, and recorded the electrophysiological activity of the transplanted GABA receptors. We found an age-dependent reduction of GABA currents in the AD brain. This reduction was larger when the AD membranes were obtained from younger subjects. We also found that GABA currents from AD brains have a faster rate of desensitization than those from non-AD brains. Furthermore, GABA receptors from AD brains were slightly, but significantly, less sensitive to GABA than receptors from non-AD brains. The reduction of GABA currents in AD was associated with reductions of mRNA and protein of the principal GABA receptor subunits normally present in the temporal cortex. Pairwise analysis of the transcripts within control and AD groups and analyses of the proportion of GABA receptor subunits revealed down-regulation of α1 and γ2 subunits in AD. In contrast, the proportions of α2, β1, and γ1 transcripts were up-regulated in the AD brains. Our data support a functional remodeling of GABAergic neurotransmission in the human AD brain.
Collapse
|
18
|
Iwakiri M, Mizukami K, Ikonomovic MD, Ishikawa M, Abrahamson EE, DeKosky ST, Asada T. An immunohistochemical study of GABA A receptor gamma subunits in Alzheimer's disease hippocampus: relationship to neurofibrillary tangle progression. Neuropathology 2009; 29:263-9. [PMID: 19019179 PMCID: PMC3078755 DOI: 10.1111/j.1440-1789.2008.00978.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Immunohistochemical characterization of the distribution of GABA(A) receptor subunits gamma1/3 and 2 in the hippocampus relative to neurofibrillary tangle (NFT) pathology staging was performed in cognitively normal subjects (Braak stage I/II, n = 4) and two groups of Alzheimer's disease (AD) patients (Braak stage III/IV, n = 4; Braak stage V/VI, n = 8). In both Braak groups of AD patients, neuronal gamma1/3 and gamma2 immunoreactivity was preserved in all hippocampal subfields. However, compared to normal controls neuronal gamma1/3 immunoreactivity was more intense in several end-stage AD subjects. Despite increased NFT pathology in the Braak V/VI AD group, GABA(A)gamma1/3 and gamma2 immunoreactivity did not co-localize with markers of NFT. These results suggest that upregulating or preserving GABA(A)gamma1/3 and gamma2 receptors may protect neurons against neurofibrillary pathology in AD.
Collapse
Affiliation(s)
- Masahiko Iwakiri
- Department of Psychiatry, Ishizaki Hosipital,, 4698Kamiishizaki, Ibaraki-machi, Ibaraki 311-3122, Japan
| | - Katsuyoshi Mizukami
- Department of Psychiatry, Institute of Clinical Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba city, Ibaraki 305-8575, Japan
| | - Milos D. Ikonomovic
- Department of Neurology, University of Pittsburgh, 3471 Fifth Avenue, Suite 811, Pittsburgh, Pennsylvania 15213, USA
- Department of Psychiatry, University of Pittsburgh, 3471 Fifth Avenue, Suite 811, Pittsburgh, Pennsylvania 15213, USA
| | - Masanori Ishikawa
- Department of Psychiatry, National Center of Neurology and Psychiatry,4-1-1Ogawahigashi-machi, Kodaira city, Tokyo, Japan
| | - Eric E. Abrahamson
- Department of Neurology, University of Pittsburgh, 3471 Fifth Avenue, Suite 811, Pittsburgh, Pennsylvania 15213, USA
| | - Steven T. DeKosky
- Department of Neurology, University of Pittsburgh, 3471 Fifth Avenue, Suite 811, Pittsburgh, Pennsylvania 15213, USA
- Department of Psychiatry, University of Pittsburgh, 3471 Fifth Avenue, Suite 811, Pittsburgh, Pennsylvania 15213, USA
| | - Takashi Asada
- Department of Psychiatry, Institute of Clinical Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba city, Ibaraki 305-8575, Japan
| |
Collapse
|
19
|
Buckingham SD, Jones AK, Brown LA, Sattelle DB. Nicotinic acetylcholine receptor signalling: roles in Alzheimer's disease and amyloid neuroprotection. Pharmacol Rev 2009; 61:39-61. [PMID: 19293145 PMCID: PMC2830120 DOI: 10.1124/pr.108.000562] [Citation(s) in RCA: 214] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD), the major contributor to dementia in the elderly, involves accumulation in the brain of extracellular plaques containing the beta-amyloid protein (Abeta) and intracellular neurofibrillary tangles of hyperphosphorylated tau protein. AD is also characterized by a loss of neurons, particularly those expressing nicotinic acetylcholine receptors (nAChRs), thereby leading to a reduction in nAChR numbers. The Abeta(1-42) protein, which is toxic to neurons, is critical to the onset and progression of AD. The discovery of new drug therapies for AD is likely to be accelerated by an improved understanding of the mechanisms whereby Abeta causes neuronal death. We examine the evidence for a role in Abeta(1-42) toxicity of nAChRs; paradoxically, nAChRs can also protect neurons when activated by nicotinic ligands. Abeta peptides and nicotine differentially activate several intracellular signaling pathways, including the phosphatidylinositol 3-kinase/v-akt murine thymoma viral oncogene homolog pathway, the extracellular signal-regulated kinase/mitogen-activated protein kinase, and JAK-2/STAT-3 pathways. These pathways control cell death or survival and the secretion of Abeta peptides. We propose that understanding the differential activation of these pathways by nicotine and/or Abeta(1-42) may offer the prospect of new routes to therapy for AD.
Collapse
Affiliation(s)
- Steven D Buckingham
- Medical Research Council Functional Genomics Unit, Department of Physiology Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX
| | | | | | | |
Collapse
|
20
|
Abstract
In this article we present a comprehensive review of relevant research and reports on the GABA(A) receptor in the aged and Alzheimer's disease (AD) brain. In comparison to glutamatergic and cholinergic systems, the GABAergic system is relatively spared in AD, but the precise mechanisms underlying differential vulnerability are not well understood. Using several methods, investigations demonstrate that despite resistance of the GABAergic system to neurodegeneration, particular subunits of the GABA(A) receptor are altered with age and AD, which can induce compensatory increases in GABA(A) receptor subunits within surrounding cells. We conclude that although aging- and disease-related changes in GABA(A) receptor subunits may be modest, the mechanisms that compensate for these changes may alter the pharmacokinetic and physiological properties of the receptor. It is therefore crucial to understand the subunit composition of individual GABA(A) receptors in the diseased brain when developing therapeutics that act at these receptors.
Collapse
Affiliation(s)
- Robert A Rissman
- Laboratory of Neuronal Structure and Function, The Salk Institute for Biological Studies, La Jolla, California, USA.
| | | | | |
Collapse
|
21
|
Abstract
As part of the hippocampus, the dentate gyrus is considered to play a crucial role in associative memory. The reviewed data suggest that the dentate gyrus withstands the formation of plaques, tangles and neuronal death until late stages of Alzheimer's disease (AD). However, changes related to a disconnecting process, and more subtle intrinsic alterations, may contribute to disturbances in memory and learning observed in early stages of AD.
Collapse
Affiliation(s)
- Thomas G Ohm
- Institute of Integrative Neuroanatomy, Department of Clinical Cell and Neurobiology, Charité CCM, 10098 Berlin, Germany.
| |
Collapse
|
22
|
Tateishi N, Shimoda T, Manako JI, Katsumata S, Shinagawa R, Ohno H. Relevance of astrocytic activation to reductions of astrocytic GABAA receptors. Brain Res 2006; 1089:79-91. [PMID: 16643860 DOI: 10.1016/j.brainres.2006.02.139] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2005] [Revised: 02/15/2006] [Accepted: 02/24/2006] [Indexed: 11/22/2022]
Abstract
Although astrocytes express gamma-aminobutyric acid subtype-A (GABAA) receptors in the mature brain, GABAA receptor expression in a cultivation state remains controversial. In this study, we investigated the alteration of astrocytic GABAA receptor expression in in vitro and in vivo studies to elucidate the relevance of astrocytic activation to reductions of astrocytic GABAA receptors. The GABA-evoked Cl- current (GABAA response) in cultured astrocytes was determined by recording in the whole-cell mode using a conventional patch-clamp technique under voltage-clamp conditions. The respective amplitudes of GABAA responses on days in vitro 1, 3-5, 7-10, and 12-15 were 1019+/-97, 512+/-76, 84+/-21, and 22+/-9 pA, respectively, suggesting that the GABAA response subsequently diminished with in vitro aging. In immunohistochemical and biochemical analyses, the expression of GABAA receptor beta-subunit decreased, whereas expressions of glial fibrillary acidic protein (GFAP) and S100B, hallmarks of astrocytic activation, increased dramatically in the cultured astrocytes with in vitro aging. With the use of [3H]SR95531, a GABAA-specific ligand, at 24 h after transient focal ischemia, binding was significantly reduced in the astrocytic fractions without affecting the synaptosomal fractions, and decreases in the mRNA expression level of GABAA receptor beta-subunits were concurrently observed. Interestingly, the loss of GABAA response in cultured astrocytes was mitigated by co-culturing with neurons or treatments with monoclonal S100B antibodies. These results indicate that astrocytic GABAA receptors are reduced with in vitro aging and cerebral ischemia, presumably through the overproduction of S100B in activated astrocytes.
Collapse
Affiliation(s)
- Narito Tateishi
- Minase Research Institute, Ono Pharmaceutical Co., Ltd., 3-1-1 Sakurai, Shimamoto-cho, Mishima-gun, Osaka 618-8585, Japan.
| | | | | | | | | | | |
Collapse
|
23
|
Rissman RA, Nocera R, Fuller LM, Kordower JH, Armstrong DM. Age-related alterations in GABAA receptor subunits in the nonhuman primate hippocampus. Brain Res 2006; 1073-1074:120-30. [PMID: 16430870 DOI: 10.1016/j.brainres.2005.12.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2005] [Revised: 11/23/2005] [Accepted: 12/05/2005] [Indexed: 10/25/2022]
Abstract
Pharmacological studies have documented that altered drug responses, particularly to benzodiazepines, are common in elderly populations. While numerous factors may contribute to changes in drug response, age-related alterations in the molecular composition of GABA(A) receptors may be a key factor in regulating these responses. We employed quantitative densitometry to examine the cytological features and density of highly prevalent hippocampal GABA(A) receptor subunits (alpha1 and beta2/3) in young and aged rhesus monkeys. alpha1 and beta2/3 subunit immunostaining was differentially distributed throughout the hippocampus. In addition, beta2/3 immunolabeling in aged monkeys was characterized by marked intersubject variability in labeling intensity, with dramatic reductions present in 3 of 5 samples. alpha1 immunolabeling in aged monkeys was significantly reduced in the CA2 and CA3 subregions, and in hilus/polymorphic layer of the dentate gyrus. Collectively, our findings demonstrate that not only are GABA(A) receptor subunits differentially distributed throughout the hippocampus, but they are also differentially altered with increased age--changes that may have an important impact on the binding properties of GABA(A) receptor pharmacological agents.
Collapse
Affiliation(s)
- Robert A Rissman
- Department of Neurobiology and Anatomy, Graduate Program in Neuroscience, MCP Hahnemann University School of Medicine, Philadelphia, PA 19102-1192, USA.
| | | | | | | | | |
Collapse
|
24
|
Dean B, Scarr E, McLeod M. Changes in hippocampal GABAA receptor subunit composition in bipolar 1 disorder. ACTA ACUST UNITED AC 2005; 138:145-55. [PMID: 15950312 DOI: 10.1016/j.molbrainres.2005.04.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2004] [Revised: 03/11/2005] [Accepted: 04/17/2005] [Indexed: 01/28/2023]
Abstract
Postmortem CNS studies have suggested an uncoupling of the gamma-aminobutyric acid (GABA) and benzodiazepine binding sites on the hippocampal GABA(A) receptor in schizophrenia. The GABA(A) receptor is an assembly of discrete subunits that form a ligand-gated ion channel, the binding characteristics of which are defined by receptor subunit composition. Thus, a likely explanation for an uncoupling between the GABA and benzodiazepine binding sites on the GABA(A) receptor would be a change in receptor subunit composition. To test this hypothesis we measured the density of GABA ([(3)H]muscimol) and benzodiazepine ([(3)H]flumazenil) binding sites on the GABA(A) receptor in hippocampi, obtained postmortem, from schizophrenic, bipolar I disorder and control subjects. In addition, we measured the amount of [(3)H]flumazenil binding that could be displaced with zolpidem and clonazepam. Levels of both [(3)H]muscimol and [(3)H]flumazenil binding were significantly decreased in part of the CA2 from subjects with schizophrenia; the decrease in [(3)H]flumazenil being due to decreases in both zolpidem-sensitive and -insensitive radioligand binding. There were complex regionally specific changes in [(3)H]muscimol binding in the hippocampus from subjects with bipolar I disorder but there were no significant changes in the overall levels of [(3)H]flumazenil binding. There were significant decreases in zolpidem-sensitive and increases in zolpidem-insensitive [(3)H]flumazenil binding in most regions of the sections of the hippocampal formation studied in bipolar I disorder. Unlike [(3)H]flumazenil, zolpidem does not bind to the alpha5 subunit of the GABA(A) receptor; therefore, we postulate that there is an increase in GABA(A) receptors containing alpha5 subunit in the hippocampus from subjects with bipolar I disorder.
Collapse
Affiliation(s)
- Brian Dean
- The Rebecca L. Cooper Research Laboratories, The Mental Health Research Institute of Victoria, Parkville, Victoria, Australia.
| | | | | |
Collapse
|
25
|
Iwakiri M, Mizukami K, Ikonomovic MD, Ishikawa M, Hidaka S, Abrahamson EE, DeKosky ST, Asada T. Changes in hippocampal GABABR1 subunit expression in Alzheimer's patients: association with Braak staging. Acta Neuropathol 2005; 109:467-74. [PMID: 15759131 DOI: 10.1007/s00401-005-0985-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2004] [Revised: 12/30/2004] [Accepted: 01/03/2005] [Indexed: 11/27/2022]
Abstract
Alterations in the gamma-aminobutyric acid (GABA) neurotransmitter and receptor systems may contribute to vulnerability of hippocampal pyramidal neurons in Alzheimer's disease (AD). The present study examined the immunohistochemical localization and distribution of GABA(B) receptor R1 protein (GBR1) in the hippocampus of 16 aged subjects with a range of neurofibrillary tangle (NFT) pathology as defined by Braak staging (I-VI). GBR1 immunoreactivity (IR) was localized to the soma and processes of hippocampal pyramidal cells and some non-pyramidal interneurons. In control subjects (Braak I/II), the intensity of neuronal GBR1 immunostaining differed among hippocampal fields, being most prominent in the CA4 and CA3/2 fields, moderate in the CA1 field, and very light in the dentate gyrus. AD cases with moderate NFT pathology (Braak III/IV) were characterized by increased GBR1-IR, particularly in the CA4 and CA3/2 fields. In the CA1 field of the majority of AD cases, the numbers of GBR1-IR neurons were significantly reduced, despite the presence of Nissl-labeled neurons in this region. These data indicate that GBR1 expression changes with the progression of NFT in AD hippocampus. At the onset of hippocampal pathology, increased or stable expression of GBR1 could contribute to neuronal resistance to the disease process. Advanced hippocampal pathology appears to be associated with decreased neuronal GBR1 staining in the CA1 region, which precedes neuronal cell death. Thus, changes in hippocampal GBR1 may reflect alterations in the balance between excitatory and inhibitory neurotransmitter systems, which likely contributes to dysfunction of hippocampal circuitry in AD.
Collapse
Affiliation(s)
- Masahiko Iwakiri
- Department of Psychiatry, Institute of Clinical Medicine, University of Tsukuba, 1-1-1 Tennodai, 305-8575, Tsukuba city, Ibaraki, Japan
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Rissman RA, Bennett DA, Armstrong DM. Subregional analysis of GABAA receptor subunit mRNAs in the hippocampus of older persons with and without cognitive impairment. J Chem Neuroanat 2004; 28:17-25. [PMID: 15363487 DOI: 10.1016/j.jchemneu.2004.05.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2004] [Revised: 05/11/2004] [Accepted: 05/12/2004] [Indexed: 11/15/2022]
Abstract
We employed in situ hybridization and quantitative densitometry techniques to examine hippocampal mRNA expression of GABA(A) receptor subunits alpha1 and alpha5 in human subjects with progressing cognitive impairment. Included in this study were 17 participants of the Religious Order Study (ROS), who were categorized into three groups based upon degree of cognitive impairment: no cognitive impairment (n = 6); moderate cognitive impairment (n = 5); and probable Alzheimer's disease (AD) (n = 6). While the levels of each specific subunit mRNA were relatively homogeneously distributed throughout the five hippocampal subregions analyzed (CA1-4, and the granule cell layer of the dentate gyrus), mRNA expression of the alpha1 receptor subunit was found to be 20% reduced in the moderate cognitive impairment group as compared to the no cognitive impairment group. In addition, alpha1 mRNA expression was 25% reduced in the probable Alzheimer's disease group compared to the group with no cognitive impairment. Similarly, alpha5 subunit mRNA was reduced 32% between no cognitive impairment and moderate cognitive impairment groups, and 35% between no cognitive impairment and probable Alzheimer's disease groups. No significant reductions were found between moderate cognitive impairment and probable Alzheimer's disease groups for either subunit. Collectively, our data provide evidence for modest reductions in GABA(A) receptor subunit mRNAs, and suggest these changes occur very early in the progression of Alzheimer's disease cognitive impairment.
Collapse
Affiliation(s)
- Robert A Rissman
- Department of Neurobiology and Anatomy, Graduate Program in Neuroscience, MCP Hahnemann University School of Medicine, Philadelphia, PA, USA.
| | | | | |
Collapse
|
27
|
Steiger JL, Russek SJ. GABAA receptors: building the bridge between subunit mRNAs, their promoters, and cognate transcription factors. Pharmacol Ther 2004; 101:259-81. [PMID: 15031002 DOI: 10.1016/j.pharmthera.2003.12.002] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The type A gamma-aminobutyric acid (GABA(A)) receptors mediate the majority of fast inhibitory neurotransmission in the CNS, and alterations in GABA(A) receptor function is believed to be involved in the pathology of several neurological and psychiatric illnesses, such as epilepsy, anxiety, Alzheimer's disease, and schizophrenia. GABA(A) receptors can be assembled from eight distinct subunit families defined by sequence similarity: alpha(1-6), beta(1-3), gamma(1-3), delta, pi, theta, and rho(1-3). The regulation of GABA(A) receptor function in the brain is a highly compensating system, influencing both the number and the composition of receptors at the cell surface. While transcriptional and translational points of control operate in parallel, it is becoming increasingly evident that many functional changes in GABA(A) receptors reflect the differential gene regulation of its subunits. The fact that certain GABA(A) receptor subunit genes are transcribed in distinct cell types during specific periods of development strongly suggests that genetic control plays a major role in the choice of subunit variants available for receptor assembly. This review focuses on the physiological conditions that alter subunit mRNA levels, the promoters that may control such levels, and the use of a conceptual framework created by bioinformatics to study coordinate and independent GABA(A) receptor subunit gene regulation. As this exciting field moves closer to identifying the language hidden inside the chromatin of GABA(A) receptor subunit gene clusters, future experiments will be aimed at testing models generated by computational analysis with biologically relevant in vivo and in vitro assays. It is hoped that through this functional genomic approach there will be the identification of new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Janine L Steiger
- Laboratory of Molecular Neurobiology, Department of Pharmacology, Boston University School of Medicine, Boston, MA 02118, USA
| | | |
Collapse
|
28
|
Carter TL, Rissman RA, Mishizen-Eberz AJ, Wolfe BB, Hamilton RL, Gandy S, Armstrong DM. Differential preservation of AMPA receptor subunits in the hippocampi of Alzheimer's disease patients according to Braak stage. Exp Neurol 2004; 187:299-309. [PMID: 15144856 DOI: 10.1016/j.expneurol.2003.12.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2003] [Accepted: 12/30/2003] [Indexed: 01/30/2023]
Abstract
The Alzheimer's disease (AD) brain, characterized pathologically by the presence of senile plaques and neurofibrillary tangles, contains regions that are differentially prone toward development of AD pathology. Within these "vulnerable" regions, specific cell populations appear to be selectively affected; the pyramidal cells of the hippocampal subiculum subfield constitute such a vulnerable region. This study investigated whether the AMPA receptor subunit content (GluR1, GluR2, GluR2/3) within "vulnerable" vs. "resistant" sectors of the hippocampus is quantitatively altered with increasing AD neuropathology, as determined by Braak staging. We hypothesize that the glutamate-mediated vulnerability is highly influenced by the repertoire of glutamate receptors expressed on hippocampal neurons. Our results indicate that AMPA receptor subunit proteins are relatively spared across all Braak stages in resistant subfields (CA2/CA3/Dentate Gyrus). However, within vulnerable sectors, i.e., subiculum, GluR2, and GluR2/3 protein levels decreased 63.77% and 60.60%, respectively, in association with Braak stages I-II and stages III-IV, respectively. In Braak stages V-VI, GluR2 and GluR2/3 protein levels were similar to those of Braak stages I-II. In contrast to GluR2 and GluR2/3, GluR1 protein levels were unchanged within vulnerable sectors throughout all stages of the disease. In interpreting these data, it may be relevant to consider that the GluR2 subunit impedes the flow of Ca(+2) through the AMPA receptor ion channel. Thus, we hypothesize that in resistant sectors, the presence of the GluR2 subunit may provide a neuroprotective role by limiting the flow of extracellular Ca(+2), whereas in vulnerable regions, the reduction of GluR2 may contribute to the vulnerability via a mechanism involving an increase in intracellular Ca(+2) and destabilization of intracellular Ca(+2) homeostasis.
Collapse
Affiliation(s)
- Troy L Carter
- Laboratory of Neuronal Vulnerability and Aging, The Lankenau Institute for Medical Research, Jefferson Health System, Wynewood, PA 19096, USA.
| | | | | | | | | | | | | |
Collapse
|
29
|
Miledi R, Dueñas Z, Martinez-Torres A, Kawas CH, Eusebi F. Microtransplantation of functional receptors and channels from the Alzheimer's brain to frog oocytes. Proc Natl Acad Sci U S A 2004; 101:1760-3. [PMID: 14749517 PMCID: PMC341849 DOI: 10.1073/pnas.0308224100] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
About a decade ago, cell membranes from the electric organ of Torpedo and from the rat brain were transplanted to frog oocytes, which thus acquired functional Torpedo and rat neurotransmitter receptors. Nevertheless, the great potential that this method has for studying human diseases has remained virtually untapped. Here, we show that cell membranes from the postmortem brains of humans that suffered Alzheimer's disease can be microtransplanted to the plasma membrane of Xenopus oocytes. We show also that these postmortem membranes carry neurotransmitter receptors and voltage-operated channels that are still functional, even after they have been kept frozen for many years. This method provides a new and powerful approach to study directly the functional characteristics and structure of receptors, channels, and other membrane proteins of the Alzheimer's brain. This knowledge may help in understanding the basis of Alzheimer's disease and also help in developing new treatments.
Collapse
Affiliation(s)
- R Miledi
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697-4550, USA.
| | | | | | | | | |
Collapse
|
30
|
Mishizen-Eberz AJ, Rissman RA, Carter TL, Ikonomovic MD, Wolfe BB, Armstrong DM. Biochemical and molecular studies of NMDA receptor subunits NR1/2A/2B in hippocampal subregions throughout progression of Alzheimer's disease pathology. Neurobiol Dis 2004; 15:80-92. [PMID: 14751773 DOI: 10.1016/j.nbd.2003.09.016] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by loss of specific cell populations within selective subregions of the hippocampus. Excitotoxicity, mediated via ionotropic glutamate receptors, may play a crucial role in this selective neuronal vulnerability. We investigated whether alterations in NMDA receptor subunits occurred during AD progression. Employing biochemical and in situ hybridization techniques in subjects with a broad range of AD pathology, protein levels, and mRNA expression of NR1/2A/2B subunits were assayed. With increasing AD neuropathology, protein levels and mRNA expression for NR1/2B subunits were significantly reduced, while the NR2A subunit mRNA expression and protein levels were unchanged. Cellular analysis of neuronal mRNA expression revealed a significant increase in the NR2A subunit in subjects with moderate neurofibrillary tangle neuropathology. This investigation supports the hypothesis that alterations occur in the expression of specific NMDA receptor subunits with increasing AD pathologic severity, which is hypothesized to contribute to the vulnerability of these neurons.
Collapse
Affiliation(s)
- Amanda J Mishizen-Eberz
- Laboratory of Neuronal Vulnerability and Aging, The Lankenau Institute for Medical Research, Jefferson Health System, Wynnewood, PA, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Armstrong DM, Sheffield R, Mishizen-Eberz AJ, Carter TL, Rissman RA, Mizukami K, Ikonomovic MD. Plasticity of glutamate and GABAA receptors in the hippocampus of patients with Alzheimer's disease. Cell Mol Neurobiol 2003; 23:491-505. [PMID: 14514010 PMCID: PMC11530205 DOI: 10.1023/a:1025063811290] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
AIM In Alzheimer's disease (AD) it is well known that specific regions of the brain are particularly vulnerable to the pathologic insults of the disease. In particular, the hippocampus is affected very early in the disease and by end stage AD is ravaged by neurofibrillary tangles and senile plaques (i.e., the pathologic hallmarks of AD). Throughout the past several years our laboratory has sought to determine the molecular mechanisms underlying the selective vulnerability of neurons in AD. METHODS To this end, we employed immunohistochemical, biochemical, and in situ hybrization methods to examine glutamate and gamma-aminobutyric acid (GABAA) receptor subtypes in the hippocampus of patients displaying the full spectrum of AD pathology. RESULTS Despite the fact that the hippocampus is characterized by a marked loss of neurons in the late stages of the disease, our data demonstrate a rather remarkable preservation among some glutamate and GABAA receptor subtypes. CONCLUSIONS Collectively, our data support the view that the relatively constant levels of selected receptor subtypes represent a compensatory up-regulation of these receptors subunits in surviving neurons. The demonstration that glutamate and GABA receptor subunits are comparably unaffected implies that even in the terminal stages of the discase the brain is "attempting" to maintain a balance in excitatory and inhibitory tone. Our data also support the concept that receptor subunits are differentially affected in AD with some subunits displaying no change while others display alterations in protein and mRNA levels within selected regions of the hippocampus. Although many of these changes are modest, they do suggest that the subunit composition of these receptors may be altered and hence affect the pharmacokinetic and physiological properties of the receptor. The latter findings stress the importance of understanding the subunit composition of individual glutamate/GABA receptors in the diseased brain prior to the development of drugs targeted towards those receptors.
Collapse
Affiliation(s)
- David M Armstrong
- Laboratory of Neuronal Vulnerability and Aging, The Lankenau Institute for Medical Research, Jefferson Health System, Wynnewood, Pennsylvania, USA.
| | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Alzheimer's disease (AD) is the most common age-related neurodegenerative disorder. Behavioural, cognitive and memory dysfunctions are characteristic symptoms of AD. The formation of amyloid plaques is currently considered as the key event of AD. Other histological hallmarks of the disease are the formation of fibrillary tangles, astrocytosis, and loss of certain neuronal systems in cortical areas of the brain. A great number of possible aetiologic and pathogenetic factors of AD have been published in the course of the last two decades. Among the toxic factors, which have been considered to contribute to the symptoms and progression of AD, ammonia deserves special interest for the following reasons: (a) Ammonia is formed in nearly all tissues and organs of the vertebrate organism; it is the most common endogenous neurotoxic compounds. Its effects on glutamatergic and GABAergic neuronal systems, the two prevailing neuronal systems of the cortical structures, are known for many years. (b) The impairment of ammonia detoxification invariably leads to severe pathology. Several symptoms and histologic aberrations of hepatic encephalopathy (HE), of which ammonia has been recognised as a pathogenetic factor, resemble those of AD. (c) The excessive formation of ammonia in the brains of AD patients has been demonstrated, and it has been shown that some AD patients exhibit elevated blood ammonia concentrations. (d) There is evidence for the involvement of aberrant lysosomal processing of beta-amyloid precursor protein (beta-APP) in the formation of amyloid deposits. Ammonia is the most important natural modulator of lysosomal protein processing. (e) Inflammatory processes and activation of microglia are widely believed to be implicated in the pathology of AD. Ammonia is able to affect the characteristic functions of microglia, such as endocytosis, and cytokine production. Based on these facts, an ammonia hypothesis of AD has first been suggested in 1993. In the present review old and new observations are discussed, which are in support of the notion that ammonia is a factor able to produce symptoms of AD and to affect the progression of the disease.
Collapse
Affiliation(s)
- Nikolaus Seiler
- Laboratory of Nutritional Oncology, Institut de Recherche Contre les Cancers de l'Appareil Digestif, Strasbourg, France.
| |
Collapse
|
33
|
Howell O, Atack JR, Dewar D, McKernan RM, Sur C. Density and pharmacology of alpha5 subunit-containing GABA(A) receptors are preserved in hippocampus of Alzheimer's disease patients. Neuroscience 2000; 98:669-75. [PMID: 10891610 DOI: 10.1016/s0306-4522(00)00163-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The anatomical localization and pharmacology of alpha5 subunit-containing GABA type-A receptors in the human hippocampal formation of Alzheimer's disease patients were studied with an alpha5 receptor selective ligand, [3H]L-655,708 and compared to age-matched human controls. Autoradiographic analyses revealed a heterogeneous distribution of [3H]L-655,708 binding sites in CA1-CA3 areas with high levels in stratum oriens, stratum pyramidale and stratum radiatum contrasting with low levels in stratum lacunosum. The highest quantity of alpha5 receptors was found in the molecular layer of the dentate gyrus. This pattern of expression was identical in both hippocampus from control and Alzheimer's disease subjects. Quantitative studies demonstrated that the number of [3H]L-655,708 binding sites is well preserved in Alzheimer's disease with only a moderate reduction (25-30%) in the CA1 subfield and entorhinal cortex. Furthermore, saturation and competition experiments with [3H]L-655,708 and representative benzodiazepine site ligands revealed that alpha5 receptors in Alzheimer's hippocampus have an alpha5beta2/3gamma2 pharmacology and structure as in control human brain.Overall, the data reported here provide evidence for a specific expression and relative sparing of alpha5 subunit-containing gamma-aminobutyric acid type-A receptors in the hippocampus of Alzheimer's patients.
Collapse
Affiliation(s)
- O Howell
- Department of Biochemistry, Merck Sharp and Dohme Research Laboratories, Terlings Park, Eastwick Road, Essex CM20 2QR, Harlow, UK
| | | | | | | | | |
Collapse
|