51
|
Lithgow BJ, Dastgheib Z, Moussavi Z. Baseline Prediction of rTMS efficacy in Alzheimer patients. Psychiatry Res 2022; 308:114348. [PMID: 34952254 DOI: 10.1016/j.psychres.2021.114348] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/13/2021] [Accepted: 12/16/2021] [Indexed: 10/19/2022]
Abstract
Repetitive transcranial magnetic stimulation (rTMS) with extensive 2-6-week protocols are applied to improve cognition and/or slow the cognitive decline seen in Alzheimer's Disease (AD). To date, there are no means to predict the response of a patient to rTMS treatment at baseline. Electrovestibulography (EVestG) biomarkers can be used to predict, at baseline, the efficacy of rTMS when applied to AD individuals. In a population of 27 AD patients (8 with significant cerebrovascular symptomatology, labelled ADcvd) EVestG signals were measured before and after rTMS treatment, and then compared with 16 age-matched healthy controls. MoCA was measured at baseline, whilst ADAS-Cog was the primary outcome measure. AD severity and comorbid cerebrovascular disease were treated as covariates. Using ADAS-Cog total score change, 13/27 AD/ADcvd patients improved with rTMS and 14/27 showed no-improvement. Leave-one-out-cross-validated linear-discriminant-analysis using two EVestG features yielded a blind accuracy of 75% for separating the improved and non-improved populations. Three-way separation of improved/non-improved/control accuracy was 91.9% using MoCA (67% alone) and one EVestG feature (66% alone). AD severity affects the rTMS treatment efficacy. The effect of existing significant cerebrovascular symptomatology on the efficacy of rTMS treatment remains unresolved. Baseline EVestG features can be predictive of the efficacy of rTMS treatment.
Collapse
Affiliation(s)
- Brian J Lithgow
- Diagnostic and Neurosignal Processing Research Laboratory, Biomedical Engineering Program, University of Manitoba, Riverview Health Centre, Manitoba, Canada; Monash Alfred Psychiatry Research Centre, Victoria, Australia.
| | - Zeinab Dastgheib
- Diagnostic and Neurosignal Processing Research Laboratory, Biomedical Engineering Program, University of Manitoba, Riverview Health Centre, Manitoba, Canada
| | - Zahra Moussavi
- Diagnostic and Neurosignal Processing Research Laboratory, Biomedical Engineering Program, University of Manitoba, Riverview Health Centre, Manitoba, Canada
| |
Collapse
|
52
|
Haghighijoo Z, Zamani L, Moosavi F, Emami S. Therapeutic potential of quinazoline derivatives for Alzheimer's disease: A comprehensive review. Eur J Med Chem 2022; 227:113949. [PMID: 34742016 DOI: 10.1016/j.ejmech.2021.113949] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/02/2021] [Accepted: 10/22/2021] [Indexed: 12/24/2022]
Abstract
Quinazolines are considered as a promising class of bioactive heterocyclic compounds with broad properties. Particularly, the quinazoline scaffold has an impressive role in the design and synthesis of new CNS-active drugs. The drug-like properties and pharmacological characteristics of quinazoline could lead to different drugs with various targets. Among CNS disorders, Alzheimer's disease (AD) is a progressive neurodegenerative disorder with memory loss, cognitive decline and language dysfunction. AD is a complex and multifactorial disease therefore, the need for finding multi-target drugs against this devastative disease is urgent. A literature survey revealed that quinazoline derivatives have diverse therapeutic potential for AD as modulators/inhibitors of β-amyloid, tau protein, cholinesterases, monoamine oxidases, and phosphodiesterases as well as other protective effects. Thus, we describe here the most relevant and recent studies about anti-AD agents with quinazoline structure which can further aid the development and discovery of new anti-AD agents.
Collapse
Affiliation(s)
- Zahra Haghighijoo
- Department of Chemistry, University of Louisiana at Lafayette, Lafayette, LA, 70504, USA
| | - Leila Zamani
- Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
| | - Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Emami
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
53
|
Cutuli D, Giacovazzo G, Decandia D, Coccurello R. Alzheimer's disease and depression in the elderly: A trajectory linking gut microbiota and serotonin signaling. Front Psychiatry 2022; 13:1010169. [PMID: 36532180 PMCID: PMC9750201 DOI: 10.3389/fpsyt.2022.1010169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/28/2022] [Indexed: 12/05/2022] Open
Abstract
The occurrence of neuropsychiatric symptoms in the elderly is viewed as an early sign of subsequent cognitive deterioration and conversion from mild cognitive impairment to Alzheimer's disease. The prognosis in terms of both the severity and progression of clinical dementia is generally aggravated by the comorbidity of neuropsychiatric symptoms and decline in cognitive function. Undeniably, aging and in particular unhealthy aging, is a silent "engine of neuropathology" over which multiple changes take place, including drastic alterations of the gut microbial ecosystem. This narrative review evaluates the role of gut microbiota changes as a possible unifying concept through which the comorbidity of neuropsychiatric symptoms and Alzheimer's disease can be considered. However, since the heterogeneity of neuropsychiatric symptoms, it is improbable to describe the same type of alterations in the bacteria population observed in patients with Alzheimer's disease, as well as it is improbable that the variety of drugs used to treat neuropsychiatric symptoms might produce changes in gut bacterial diversity similar to that observed in the pathophysiology of Alzheimer's disease. Depression seems to be another very intriguing exception, as it is one of the most frequent neuropsychiatric symptoms in dementia and a mood disorder frequently associated with brain aging. Antidepressants (i.e., serotonin reuptake inhibitors) or tryptophan dietary supplementation have been shown to reduce Amyloid β-loading, reinstate microbial diversity and reduce the abundance of bacterial taxa dominant in depression and Alzheimer's disease. This review briefly examines this trajectory by discussing the dysfunction of gut microbiota composition, selected bacterial taxa, and alteration of tryptophan and serotonin metabolism/neurotransmission as overlapping in-common mechanisms involved with depression, Alzheimer's disease, and unhealthy aging.
Collapse
Affiliation(s)
- Debora Cutuli
- Department of Psychology, University of Rome La Sapienza, Rome, Italy.,European Center for Brain Research, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Giacomo Giacovazzo
- European Center for Brain Research, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Davide Decandia
- Department of Psychology, University of Rome La Sapienza, Rome, Italy.,European Center for Brain Research, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Roberto Coccurello
- European Center for Brain Research, Santa Lucia Foundation IRCCS, Rome, Italy.,Institute for Complex Systems (ISC), National Council of Research (CNR), Rome, Italy
| |
Collapse
|
54
|
Ferrer I. Alzheimer's disease is an inherent, natural part of human brain aging: an integrated perspective. FREE NEUROPATHOLOGY 2022; 3:17. [PMID: 37284149 PMCID: PMC10209894 DOI: 10.17879/freeneuropathology-2022-3806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/21/2022] [Indexed: 06/08/2023]
Abstract
Alzheimer disease is one of the most challenging demons in our society due to its very high prevalence and its clinical manifestations which cause deterioration of cognition, intelligence, and emotions - the very capacities that distinguish Homo sapiens from other animal species. Besides the personal, social, and economical costs, late stages of AD are vivid experiences for the family, relatives, friends, and general observers of the progressive ruin of an individual who turns into a being with lower mental and physical capacities than less evolved species. A human brain with healthy cognition, conscience, and emotions can succeed in dealing with most difficulties that life may pose. Without these capacities, the same person probably cannot. Due, in part, to this emotional impact, the absorbing study of AD has generated, over the years, a fascinating and complex story of theories, hypotheses, controversies, fashion swings, and passionate clashes, together with tremendous efforts and achievements geared to improve understanding of the pathogenesis and treatment of the disorder. Familal AD is rare and linked to altered genetic information associated with three genes. Sporadic AD (sAD) is much more common and multifactorial. A major point of clinical discussion has been, and still is, establishing the differences between brain aging and sAD. This is not a trivial question, as the neuropathological and molecular characteristics of normal brain aging and the first appearance of early stages of sAD-related pathology are not easily distinguishable in most individuals. Another important point is confidence in assigning responsibility for the beginning of sAD to a few triggering molecules, without considering the wide number of alterations that converge in the pathogenesis of aging and sAD. Genetic risk factors covering multiple molecular signals are increasing in number. In the same line, molecular pathways are altered at early stages of sAD pathology, currently grouped under the aegis of normal brain aging, only to increase massively at advanced stages of the process. Sporadic AD is here considered an inherent, natural part of human brain aging, which is prevalent in all humans, and variably present or not in a few individuals in other species. The progression of the process has devastating effects in a relatively low percentage of human beings eventually evolving to dementia. The continuum of brain aging and sAD implies the search for a different approach in the study of human brain aging at the first stages of the biological process, and advances in the use of new technologies aimed at slowing down the molecular defects underlying human brain aging and sAD at the outset, and transfering information and tasks to AI and coordinated devices.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona; Emeritus Researcher of the Bellvitge Institute of Biomedical Research (IDIBELL); Biomedical Research Network of Neurodegenerative Diseases (CIBERNED); Institute of Neurosciences, University of Barcelona; Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
55
|
Auta J, Locci A, Guidotti A, Davis JM, Dong H. Sex-dependent sensitivity to positive allosteric modulation of GABA action in an APP knock-in mouse model of Alzheimer's disease: Potential epigenetic regulation. CURRENT RESEARCH IN NEUROBIOLOGY 2022; 3:100025. [DOI: 10.1016/j.crneur.2021.100025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 12/15/2022] Open
|
56
|
Kuhlemann A, Beliu G, Janzen D, Petrini EM, Taban D, Helmerich DA, Doose S, Bruno M, Barberis A, Villmann C, Sauer M, Werner C. Genetic Code Expansion and Click-Chemistry Labeling to Visualize GABA-A Receptors by Super-Resolution Microscopy. Front Synaptic Neurosci 2021; 13:727406. [PMID: 34899260 PMCID: PMC8664562 DOI: 10.3389/fnsyn.2021.727406] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 11/02/2021] [Indexed: 01/15/2023] Open
Abstract
Fluorescence labeling of difficult to access protein sites, e.g., in confined compartments, requires small fluorescent labels that can be covalently tethered at well-defined positions with high efficiency. Here, we report site-specific labeling of the extracellular domain of γ-aminobutyric acid type A (GABA-A) receptor subunits by genetic code expansion (GCE) with unnatural amino acids (ncAA) combined with bioorthogonal click-chemistry labeling with tetrazine dyes in HEK-293-T cells and primary cultured neurons. After optimization of GABA-A receptor expression and labeling efficiency, most effective variants were selected for super-resolution microscopy and functionality testing by whole-cell patch clamp. Our results show that GCE with ncAA and bioorthogonal click labeling with small tetrazine dyes represents a versatile method for highly efficient site-specific fluorescence labeling of proteins in a crowded environment, e.g., extracellular protein domains in confined compartments such as the synaptic cleft.
Collapse
Affiliation(s)
- Alexander Kuhlemann
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Würzburg, Germany
| | - Gerti Beliu
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Würzburg, Germany.,Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Wuerzburg, Würzburg, Germany
| | - Dieter Janzen
- Institute of Clinical Neurobiology, University of Würzburg, Würzburg, Germany
| | - Enrica Maria Petrini
- Neuroscience and Brain Technologies Department, Istituto Italiano Di Tecnologia, Genova, Italy
| | - Danush Taban
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Würzburg, Germany
| | - Dominic A Helmerich
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Würzburg, Germany
| | - Sören Doose
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Würzburg, Germany
| | - Martina Bruno
- Neuroscience and Brain Technologies Department, Istituto Italiano Di Tecnologia, Genova, Italy
| | - Andrea Barberis
- Neuroscience and Brain Technologies Department, Istituto Italiano Di Tecnologia, Genova, Italy
| | - Carmen Villmann
- Institute of Clinical Neurobiology, University of Würzburg, Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Würzburg, Germany
| | - Christian Werner
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Würzburg, Germany
| |
Collapse
|
57
|
Bruzelius A, Kidnapillai S, Drouin-Ouellet J, Stoker T, Barker RA, Rylander Ottosson D. Reprogramming Human Adult Fibroblasts into GABAergic Interneurons. Cells 2021; 10:cells10123450. [PMID: 34943958 PMCID: PMC8699824 DOI: 10.3390/cells10123450] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/26/2021] [Accepted: 12/04/2021] [Indexed: 12/03/2022] Open
Abstract
Direct reprogramming is an appealing strategy to generate neurons from a somatic cell by forced expression of transcription factors. The generated neurons can be used for both cell replacement strategies and disease modelling. Using this technique, previous studies have shown that γ-aminobutyric acid (GABA) expressing interneurons can be generated from different cell sources, such as glia cells or fetal fibroblasts. Nevertheless, the generation of neurons from adult human fibroblasts, an easily accessible cell source to obtain patient-derived neurons, has proved to be challenging due to the intrinsic blockade of neuronal commitment. In this paper, we used an optimized protocol for adult skin fibroblast reprogramming based on RE1 Silencing Transcription Factor (REST) inhibition together with a combination of GABAergic fate determinants to convert human adult skin fibroblasts into GABAergic neurons. Our results show a successful conversion in 25 days with upregulation of neuronal gene and protein expression levels. Moreover, we identified specific gene combinations that converted fibroblasts into neurons of a GABAergic interneuronal fate. Despite the well-known difficulty in converting adult fibroblasts into functional neurons in vitro, we could detect functional maturation in the induced neurons. GABAergic interneurons have relevance for cognitive impairments and brain disorders, such as Alzheimer’s and Parkinson’s diseases, epilepsy, schizophrenia and autism spectrum disorders.
Collapse
Affiliation(s)
- Andreas Bruzelius
- Group of Regenerative Neurophysiology, Lund Stem Cell Center, Department of Experimental Medical Science, Faculty of Medicine, Lund University, 221 84 Lund, Sweden; (A.B.); (S.K.)
| | - Srisaiyini Kidnapillai
- Group of Regenerative Neurophysiology, Lund Stem Cell Center, Department of Experimental Medical Science, Faculty of Medicine, Lund University, 221 84 Lund, Sweden; (A.B.); (S.K.)
| | | | - Tom Stoker
- Wellcome-MRC Cambridge Stem Cell Institute and John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK; (T.S.); (R.A.B.)
| | - Roger A. Barker
- Wellcome-MRC Cambridge Stem Cell Institute and John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK; (T.S.); (R.A.B.)
| | - Daniella Rylander Ottosson
- Group of Regenerative Neurophysiology, Lund Stem Cell Center, Department of Experimental Medical Science, Faculty of Medicine, Lund University, 221 84 Lund, Sweden; (A.B.); (S.K.)
- Correspondence: ; Tel.: +46-222-0559
| |
Collapse
|
58
|
Role of Receptors in Relation to Plaques and Tangles in Alzheimer's Disease Pathology. Int J Mol Sci 2021; 22:ijms222312987. [PMID: 34884789 PMCID: PMC8657621 DOI: 10.3390/ijms222312987] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/26/2021] [Accepted: 11/28/2021] [Indexed: 12/23/2022] Open
Abstract
Despite the identification of Aβ plaques and NFTs as biomarkers for Alzheimer’s disease (AD) pathology, therapeutic interventions remain elusive, with neither an absolute prophylactic nor a curative medication available to impede the progression of AD presently available. Current approaches focus on symptomatic treatments to maintain AD patients’ mental stability and behavioral symptoms by decreasing neuronal degeneration; however, the complexity of AD pathology requires a wide range of therapeutic approaches for both preventive and curative treatments. In this regard, this review summarizes the role of receptors as a potential target for treating AD and focuses on the path of major receptors which are responsible for AD progression. This review gives an overall idea centering on major receptors, their agonist and antagonist and future prospects of viral mimicry in AD pathology. This article aims to provide researchers and developers a comprehensive idea about the different receptors involved in AD pathogenesis that may lead to finding a new therapeutic strategy to treat AD.
Collapse
|
59
|
Zhou Y, Cheng Y, Li Y, Ma J, Wu Z, Chen Y, Mei J, Chen M. Soluble β-amyloid impaired the GABA inhibition by mediating KCC2 in early APP/PS1 mice. Biosci Trends 2021; 15:330-340. [PMID: 34526443 DOI: 10.5582/bst.2021.01245] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, which has become the leading cause of dementia cases globally. Synaptic failure is an early pathological feature of AD. However, the cause of synaptic failure in AD, especially the GABAergic synaptic activity remains unclear. Extensive evidence indicates that the presence of soluble amyloid-β is an early pathological feature in AD, which triggers synaptic dysfunction and cognitive decline. Our recent study explored the relation of GABAergic transmission and soluble Aβ in early APP/PS1 mice. Firstly, we found soluble Aβ42 levels were significantly increased in serum, hippocampus and prefrontal cortex in 3-4 months APP/PS1 mice, which was much earlier than Aβ plagues formation. In addition, we found TNF-α and BDNF expression levels were increased, while KCC2 and GABAAR expression were decreased in 3-4 months APP/PS1 hippocampus. When we treated 3-4 months APP/PS1 mice with a potent γ-secretase inhibitor, LY411575, which can reduce the soluble Aβ42 levels, the TNF-α and BDNF protein levels were decreased, while KCC2 and GABAAR levels were increased. In conclusion, our study suggested soluble Aβ may impaired the GABA inhibition by mediating KCC2 levels in early APP/PS1 mice. KCC2 may be served as a potential biomarker for AD.
Collapse
Affiliation(s)
- Yuan Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Yujie Cheng
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Yong Li
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Jiyao Ma
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Zhihan Wu
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Yuenan Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Jinyu Mei
- Department of Otorhinolaryngology, Head and Neck Surgery, the Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ming Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
60
|
Sood A, Preeti K, Fernandes V, Khatri DK, Singh SB. Glia: A major player in glutamate-GABA dysregulation-mediated neurodegeneration. J Neurosci Res 2021; 99:3148-3189. [PMID: 34748682 DOI: 10.1002/jnr.24977] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 12/16/2022]
Abstract
The imbalance between glutamate and γ-aminobutyric acid (GABA) results in the loss of synaptic strength leading to neurodegeneration. The dogma on the field considered neurons as the main players in this excitation-inhibition (E/I) balance. However, current strategies focusing only on neurons have failed to completely understand this condition, bringing up the importance of glia as an alternative modulator for neuroinflammation as glia alter the activity of neurons and is a source of both neurotrophic and neurotoxic factors. This review's primary goal is to illustrate the role of glia over E/I balance in the central nervous system and its interaction with neurons. Rather than focusing only on the neuronal targets, we take a deeper look at glial receptors and proteins that could also be explored as drug targets, as they are early responders to neurotoxic insults. This review summarizes the neuron-glia interaction concerning GABA and glutamate, possible targets, and its involvement in the E/I imbalance in neurodegenerative diseases like Alzheimer's disease, Parkinson's disease, Huntington's disease, and multiple sclerosis.
Collapse
Affiliation(s)
- Anika Sood
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Kumari Preeti
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Valencia Fernandes
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Dharmendra Kumar Khatri
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Shashi Bala Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| |
Collapse
|
61
|
Ethiraj J, Palpagama TH, Turner C, van der Werf B, Waldvogel HJ, Faull RLM, Kwakowsky A. The effect of age and sex on the expression of GABA signaling components in the human hippocampus and entorhinal cortex. Sci Rep 2021; 11:21470. [PMID: 34728681 PMCID: PMC8563768 DOI: 10.1038/s41598-021-00792-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 10/14/2021] [Indexed: 12/02/2022] Open
Abstract
Gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the nervous system. The GABA signaling system in the brain is comprised of GABA synthesizing enzymes, transporters, GABAA and GABAB receptors (GABAAR and GABABR). Alterations in the expression of these signaling components have been observed in several brain regions throughout aging and between sexes in various animal models. The hippocampus is the memory centre of the brain and is impaired in several age-related disorders. It is composed of two main regions: the Cornu Ammonis (CA1-4) and the Dentate Gyrus (DG), which are interconnected with the Entorhinal Cortex (ECx). The age- and sex-specific changes of GABA signaling components in these regions of the human brain have not been examined. This study is the first to determine the effect of age and sex on the expression of GABA signaling components-GABAAR α1,2,3,5, β1-3, γ2, GABABR R1 and R2 subunits and the GABA synthesizing enzymes GAD 65/67-in the ECx, and the CA1 and DG regions of the human hippocampus using Western blotting. No significant differences were found in GABAAR α1,2,3,5, β1-3, γ2, GABABR R1 and R2 subunit and GAD65/76 expression levels in the ECx, CA1 and DG regions between the younger and older age groups for both sexes. However, we observed a significant negative correlation between age and GABAAR α1subunit level in the CA1 region for females; significant negative correlation between age and GABAAR β1, β3 and γ2 subunit expression in the DG region for males. In females a significant positive correlation was found between age and GABAAR γ2 subunit expression in the ECx and GABABR R2 subunit expression in the CA1 region. The results indicate that age and sex do not affect the expression of GAD 65/67. In conclusion, our results show age- and sex-related GABAA/BR subunit alterations in the ECx and hippocampus that might significantly influence GABAergic neurotransmission and underlie disease susceptibility and progression.
Collapse
Affiliation(s)
- Jayarjun Ethiraj
- grid.9654.e0000 0004 0372 3343Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Thulani Hansika Palpagama
- grid.9654.e0000 0004 0372 3343Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Clinton Turner
- grid.9654.e0000 0004 0372 3343Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, School of Medical Sciences, University of Auckland, Auckland, New Zealand ,grid.414055.10000 0000 9027 2851Department of Anatomical Pathology, LabPlus, Auckland City Hospital, Auckland, New Zealand
| | - Bert van der Werf
- grid.9654.e0000 0004 0372 3343Department of Epidemiology and Biostatistics, Faculty of Medical and Health Sciences, School of Population Health, University of Auckland, Auckland, New Zealand
| | - Henry John Waldvogel
- grid.9654.e0000 0004 0372 3343Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Richard Lewis Maxwell Faull
- grid.9654.e0000 0004 0372 3343Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Andrea Kwakowsky
- grid.9654.e0000 0004 0372 3343Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
62
|
Alavi MV. Tau phosphorylation and OPA1 proteolysis are unrelated events: Implications for Alzheimer's Disease. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2021; 1868:119116. [PMID: 34400172 PMCID: PMC8525314 DOI: 10.1016/j.bbamcr.2021.119116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/29/2021] [Accepted: 07/31/2021] [Indexed: 12/11/2022]
Abstract
The neuropathological hallmarks of Alzheimer's Disease are plaques and neurofibrillary tangles. Yet, Alzheimer's is a complex disease with many contributing factors, such as energy-metabolic changes, which have been documented in autopsy brains from individuals with Alzheimer's and animal disease models alike. One conceivable explanation is that the interplay of age-related extracellular and intracellular alterations pertaining to Alzheimer's, such as cerebrovascular changes, protein aggregates and inflammation, evoke a mitochondrial response. However, it is not clear if and how mitochondria can contribute to Alzheimer's pathophysiology. This study focuses on one particular aspect of this question by investigating the functional interaction between the microtubule-associated protein tau and the mitochondrial inner membrane fusion machinery, which shows alterations in Alzheimer's brains. OPA1 is an essential inner membrane-fusion protein regulated by the two membrane proteases OMA1 and YME1L1. Assessment of OPA1 proteolysis-usually found in dividing mitochondria-and posttranslational tau modifications in mouse and human neuroblastoma cells under different experimental conditions clarified the relationship between these two pathways: OPA1 hydrolysis and phosphorylation or dephosphorylation of tau may coincide, but are not causally related. OPA1 cleavage did not alter tau's phosphorylation pattern. Conversely, tau's phosphorylation state did not induce nor correlate with OPA1 proteolysis. These results irrefutably demonstrate that there is no direct functional interaction between posttranslational tau modifications and the regulation of the OMA1-OPA1 pathway, which implies a common root cause modulating both pathways in Alzheimer's.
Collapse
|
63
|
Czapski GA, Strosznajder JB. Glutamate and GABA in Microglia-Neuron Cross-Talk in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms222111677. [PMID: 34769106 PMCID: PMC8584169 DOI: 10.3390/ijms222111677] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 12/18/2022] Open
Abstract
The physiological balance between excitation and inhibition in the brain is significantly affected in Alzheimer’s disease (AD). Several neuroactive compounds and their signaling pathways through various types of receptors are crucial in brain homeostasis, among them glutamate and γ-aminobutyric acid (GABA). Activation of microglial receptors regulates the immunological response of these cells, which in AD could be neuroprotective or neurotoxic. The novel research approaches revealed the complexity of microglial function, including the interplay with other cells during neuroinflammation and in the AD brain. The purpose of this review is to describe the role of several proteins and multiple receptors on microglia and neurons, and their involvement in a communication network between cells that could lead to different metabolic loops and cell death/survival. Our review is focused on the role of glutamatergic, GABAergic signaling in microglia–neuronal cross-talk in AD and neuroinflammation. Moreover, the significance of AD-related neurotoxic proteins in glutamate/GABA-mediated dialogue between microglia and neurons was analyzed in search of novel targets in neuroprotection, and advanced pharmacological approaches.
Collapse
|
64
|
Xu H, Zhou Q, Liu B, Cheng KW, Chen F, Wang M. Neuroprotective Potential of Mung Bean ( Vigna radiata L.) Polyphenols in Alzheimer's Disease: A Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:11554-11571. [PMID: 34551518 DOI: 10.1021/acs.jafc.1c04049] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Mung bean contains various neuroprotective polyphenols, so it might be a healthy food for Alzheimer's disease (AD) prevention. Totally, 19 major phenolic compounds were quantified in mung bean, including 10 phenolic acids and 9 flavonoids. After summarizing their contents and effective doses in rodent AD models, it was speculated that vitexin, isovitexin, sinapic acid, and ferulic acid might be the major bioactive compounds for mung bean-mediated neuroprotection. The mechanisms involved inhibition of β-amyloidogenesis, tau hyperphosphorylation, oxidative stress, and neuroinflammation, and promotion of autophagy and acetylcholinesterase enzyme activity. Notably, the neuroprotective phenolic profile in mung bean changed after germination, with decreased vitexin and isovitexin, and increased rutin, isoquercitrin, isorhamnetin, and caffeic acid detected. However, only studies of individual phenolic compounds in mung bean are published at present. Hence, further studies are needed to elucidate the neuroprotective activities and mechanisms of extractions of mung bean seeds and sprouts, and the synergism between different phenolic compounds.
Collapse
Affiliation(s)
- Hui Xu
- College of Civil and Transportation Engineering, Shenzhen University, Shenzhen 518060, China
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China
| | - Qian Zhou
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China
| | - Bin Liu
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China
| | - Ka-Wing Cheng
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China
| | - Feng Chen
- College of Civil and Transportation Engineering, Shenzhen University, Shenzhen 518060, China
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China
| | - Mingfu Wang
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
65
|
Wander CM, Song J. The neurogenic niche in Alzheimer's disease. Neurosci Lett 2021; 762:136109. [PMID: 34271133 PMCID: PMC9013442 DOI: 10.1016/j.neulet.2021.136109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 06/17/2021] [Accepted: 07/07/2021] [Indexed: 12/15/2022]
Abstract
Adult hippocampal neurogenesis is the process of generation and functional incorporation of new neurons, formed by adult neural stem cells in the dentate gyrus. Adult hippocampal neurogenesis is highly dependent upon the integration of dynamic external stimuli and is instrumental in the formation of new spatial memories. Adult hippocampal neurogenesis is therefore uniquely sensitive to the summation of neuronal circuit and neuroimmune environments that comprise the neurogenic niche, and has powerful implications in diseases of aging and neurological disorders. This sensitivity underlies the neurogenic niche alterations commonly observed in Alzheimer's disease, the most common form of dementia. This review summarizes Alzheimer's disease associated changes in neuronal network activity, neuroinflammatory processes, and adult neural stem cell fate choice that ultimately result in neurogenic niche dysfunction and impaired adult hippocampal neurogenesis. A more comprehensive understanding of the complex changes mediating neurogenic niche disturbances in Alzheimer's disease will aid development of future therapies targeting adult neurogenesis.
Collapse
Affiliation(s)
- Connor M Wander
- Department of Pharmacology, University of North Carolina at Chapel Hill
| | - Juan Song
- Department of Pharmacology, University of North Carolina at Chapel Hill
- Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
66
|
Busquets O, Parcerisas A, Verdaguer E, Ettcheto M, Camins A, Beas-Zarate C, Castro-Torres RD, Auladell C. c-Jun N-Terminal Kinases in Alzheimer's Disease: A Possible Target for the Modulation of the Earliest Alterations. J Alzheimers Dis 2021; 82:S127-S139. [PMID: 33216036 DOI: 10.3233/jad-201053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Given the highly multifactorial origin of Alzheimer's disease (AD) neuropathology, disentangling and orderly knowing mechanisms involved in sporadic onset are arduous. Nevertheless, when the elements involved are dissected into smaller pieces, the task becomes more accessible. This review aimed to describe the link between c-Jun N-terminal Kinases (JNKs), master regulators of many cellular functions, and the early alterations of AD: synaptic loss and dysregulation of neuronal transport. Both processes have a role in the posterior cognitive decline observed in AD. The manuscript focuses on the molecular mechanisms of glutamatergic, GABA, and cholinergic synapses altered by the presence of amyloid-β aggregates and hyperphosphorylated tau, as well as on several consequences of the disruption of cellular processes linked to neuronal transport that is controlled by the JNK-JIP (c-jun NH2-terminal kinase (JNK)-interacting proteins (JIPs) complex, including the transport of AβPP or autophagosomes.
Collapse
Affiliation(s)
- Oriol Busquets
- Department of Pharmacology, Toxicology and Therapeutic Chemistry; Pharmacy and Food Sciences Faculty, Universitat de Barcelona, Barcelona, Spain.,Department of Biochemistry and Biotechnology, Medicine and Health Sciences Faculty, Universitat Rovira i Virgili, Reus, Spain.,Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Dominick P. Purpura Department of Neurosciences, Albert Einstein College of Medicine, New York City, NY, USA
| | - Antoni Parcerisas
- Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Biology Faculty, Universitat de Barcelona, Barcelona, Spain
| | - Ester Verdaguer
- Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Biology Faculty, Universitat de Barcelona, Barcelona, Spain
| | - Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry; Pharmacy and Food Sciences Faculty, Universitat de Barcelona, Barcelona, Spain.,Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry; Pharmacy and Food Sciences Faculty, Universitat de Barcelona, Barcelona, Spain.,Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Carlos Beas-Zarate
- Department of Cell and Molecular Biology, Laboratory of Neural Regeneration, C.U.C.B.A., Universidad de Guadalajara, Jalisco, Mexico
| | - Rubén Darío Castro-Torres
- Department of Cell and Molecular Biology, Laboratory of Biology of Neurotransmission, C.U.C.B.A., Universidad de Guadalajara, Jalisco, Mexico
| | - Carme Auladell
- Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Biology Faculty, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
67
|
TDP-43 regulates GAD1 mRNA splicing and GABA signaling in Drosophila CNS. Sci Rep 2021; 11:18761. [PMID: 34548578 PMCID: PMC8455590 DOI: 10.1038/s41598-021-98241-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/01/2021] [Indexed: 12/11/2022] Open
Abstract
Alterations in the function of the RNA-binding protein TDP-43 are largely associated with the pathogenesis of amyotrophic lateral sclerosis (ALS), a devastating disease of the human motor system that leads to motoneurons degeneration and reduced life expectancy by molecular mechanisms not well known. In our previous work, we found that the expression levels of the glutamic acid decarboxylase enzyme (GAD1), responsible for converting glutamate to γ-aminobutyric acid (GABA), were downregulated in TBPH-null flies and motoneurons derived from ALS patients carrying mutations in TDP-43, suggesting that defects in the regulation of GAD1 may lead to neurodegeneration by affecting neurotransmitter balance. In this study, we observed that TBPH was required for the regulation of GAD1 pre-mRNA splicing and the levels of GABA in the Drosophila central nervous system (CNS). Interestingly, we discovered that pharmacological treatments aimed to potentiate GABA neurotransmission were able to revert locomotion deficiencies in TBPH-minus flies, revealing novel mechanisms and therapeutic strategies in ALS.
Collapse
|
68
|
Monterey MD, Wei H, Wu X, Wu JQ. The Many Faces of Astrocytes in Alzheimer's Disease. Front Neurol 2021; 12:619626. [PMID: 34531807 PMCID: PMC8438135 DOI: 10.3389/fneur.2021.619626] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 07/20/2021] [Indexed: 01/11/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease and is the most common cause of dementia in an aging population. The majority of research effort has focused on the role of neurons in neurodegeneration and current therapies have limited ability to slow disease progression. Recently more attention has been given to the role of astrocytes in the process of neurodegeneration. Specifically, reactive astrocytes have both advantageous and adverse effects during neurodegeneration. The ability to isolate and depict astrocyte phenotype has been challenging. However, with the recent development of single-cell sequencing technologies researchers are provided with the resource to delineate specific biomarkers associated with reactive astrocytes in AD. In this review, we will focus on the role of astrocytes in normal conditions and the pathological development of AD. We will further review recent developments in the understanding of astrocyte heterogeneity and associated biomarkers. A better understanding of astrocyte contributions and phenotypic changes in AD can ultimately lead to more effective therapeutic targets.
Collapse
Affiliation(s)
- Michael D Monterey
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Haichao Wei
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX, United States
| | - Xizi Wu
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX, United States
| | - Jia Qian Wu
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX, United States.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| |
Collapse
|
69
|
Jia Y, Wang X, Chen Y, Qiu W, Ge W, Ma C. Proteomic and Transcriptomic Analyses Reveal Pathological Changes in the Entorhinal Cortex Region that Correlate Well with Dysregulation of Ion Transport in Patients with Alzheimer's Disease. Mol Neurobiol 2021; 58:4007-4027. [PMID: 33904022 DOI: 10.1007/s12035-021-02356-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/10/2021] [Indexed: 01/17/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder. The earliest neuropathology of AD appears in entorhinal cortex (EC) regions. Therapeutic strategies and preventive measures to protect against entorhinal degeneration would be of substantial value in the early stages of AD. In this study, transcriptome based on the Illumina RNA-seq and proteome based on TMT-labelling were performed for RNA and protein profiling on AD EC samples and non-AD control EC samples. Immunohistochemistry was used to validate proteins expressions. After integrated analysis, 57 genes were detected both in transcriptome and proteome data, including 51 in similar altering trends (7 upregulated, 44 downregulated) and 6 in inverse trends when compared AD vs. control. The top 6 genes (GABRG2, CACNG3, CACNB4, GABRB2, GRIK2, and SLC17A6) within the 51 genes were selected and related to "ion transport". Correlation analysis demonstrated negative relationship of protein expression level with the neuropathologic changes. In conclusion, the integrate transcriptome and proteome analysis provided evidence for dysregulation of ion transport across brain regions in AD, which might be a critical signaling pathway that initiates pathology. This study might provide new insight into the earliest changes occurring in the EC of AD and novel targets for AD prevention and treatment.
Collapse
Affiliation(s)
- Yangjie Jia
- Department of Human Anatomy, Histology and Embryology, Neuroscience Center, National Human Brain Bank for Development and Function, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 5 Dongdansantiao, Dongcheng District, Beijing, 100005, China
| | - Xia Wang
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 5 Dongdansantiao, Dongcheng District, Beijing, 100005, China
| | - Yanyu Chen
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 5 Dongdansantiao, Dongcheng District, Beijing, 100005, China
| | - Wenying Qiu
- Department of Human Anatomy, Histology and Embryology, Neuroscience Center, National Human Brain Bank for Development and Function, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 5 Dongdansantiao, Dongcheng District, Beijing, 100005, China
| | - Wei Ge
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 5 Dongdansantiao, Dongcheng District, Beijing, 100005, China.
| | - Chao Ma
- Department of Human Anatomy, Histology and Embryology, Neuroscience Center, National Human Brain Bank for Development and Function, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 5 Dongdansantiao, Dongcheng District, Beijing, 100005, China.
| |
Collapse
|
70
|
Jiménez-Balado J, Eich TS. GABAergic dysfunction, neural network hyperactivity and memory impairments in human aging and Alzheimer's disease. Semin Cell Dev Biol 2021; 116:146-159. [PMID: 33573856 PMCID: PMC8292162 DOI: 10.1016/j.semcdb.2021.01.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/25/2021] [Accepted: 01/30/2021] [Indexed: 02/07/2023]
Abstract
In this review, we focus on the potential role of the γ-aminobutyric acidergic (GABAergic) system in age-related episodic memory impairments in humans, with a particular focus on Alzheimer's disease (AD). Well-established animal models have shown that GABA plays a central role in regulating and synchronizing neuronal signaling in the hippocampus, a brain area critical for episodic memory that undergoes early and significant morphologic and functional changes in the course of AD. Neuroimaging research in humans has documented hyperactivity in the hippocampus and losses of resting state functional connectivity in the Default Mode Network, a network that itself prominently includes the hippocampus-presaging episodic memory decline in individuals at-risk for AD. Apolipoprotein ε4, the highest genetic risk factor for AD, is associated with GABAergic dysfunction in animal models, and episodic memory impairments in humans. In combination, these findings suggest that GABA may be the linchpin in a complex system of factors that eventually leads to the principal clinical hallmark of AD: episodic memory loss. Here, we will review the current state of literature supporting this hypothesis. First, we will focus on the molecular and cellular basis of the GABAergic system and its role in memory and cognition. Next, we report the evidence of GABA dysregulations in AD and normal aging, both in animal models and human studies. Finally, we outline a model of GABAergic dysfunction based on the results of functional neuroimaging studies in humans, which have shown hippocampal hyperactivity to episodic memory tasks concurrent with and even preceding AD diagnosis, along with factors that may modulate this association.
Collapse
Affiliation(s)
- Joan Jiménez-Balado
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Teal S Eich
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
71
|
Physiological separation of Alzheimer's disease and Alzheimer's disease with significant levels of cerebrovascular symptomology and healthy controls. Med Biol Eng Comput 2021; 59:1597-1610. [PMID: 34263439 DOI: 10.1007/s11517-021-02409-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 07/04/2021] [Indexed: 01/14/2023]
Abstract
Most dementia patients with a mixed dementia (MxD) diagnosis have a mix of Alzheimer's disease (AD) and vascular dementia. Electrovestibulography (EVestG) records vestibuloacoustic afferent activity. We hypothesize EVestG recordings of AD and MxD patients are different. All patients were assessed with the Montreal cognitive assessment (MoCA) and Hachinski ischemic scale (HIS) (> 4 HIS score < 7 is representative of MxD cerebrovascular symptomology). EVestG recordings were made from 26 AD, 21 MxD and 44 healthy (control) participants. Features were derived from the EVestG recordings of the average field potential and field potential interval histogram to classify the AD, MxD and control groups. Multivariate analysis was used to test the features' significance. Using a leave-one-out cross-validated linear discriminant analysis with 3 EVestG features yielded accuracies > 80% for separating pairs of AD/MxD/control. Using the MoCA assessment and 2 EVestG features, a best accuracy of 81 to 91% depending on the classifier was obtained for the 3-way identification of AD, MxD and controls. EVestG measures provide a physiological basis for identifying AD from MxD. EVestG measures are hypothesized to be partly related to channelopathies and changes in the descending input to the vestibular periphery. Four of the five AD or MxD versus control features used had significant correlations with the MoCA. This supports assertions that the pathologic changes associated with AD impact the vestibular system and further are suggestive that the postulated physiological changes behind these features have an association with cognitive decline severity.
Collapse
|
72
|
Kong Y, Huang L, Li W, Liu X, Zhou Y, Liu C, Zhang S, Xie F, Zhang Z, Jiang D, Zhou W, Ni R, Zhang C, Sun B, Wang J, Guan Y. The Synaptic Vesicle Protein 2A Interacts With Key Pathogenic Factors in Alzheimer's Disease: Implications for Treatment. Front Cell Dev Biol 2021; 9:609908. [PMID: 34277597 PMCID: PMC8282058 DOI: 10.3389/fcell.2021.609908] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 05/24/2021] [Indexed: 11/24/2022] Open
Abstract
Alzheimer’s disease (AD), a serious neurodegenerative disease, is pathologically characterized by synaptic loss and dysfunction. Synaptic vesicle protein 2A (SV2A) is an indispensable vesicular protein specifically expressed in synapses and can be used as a biomarker for synaptic density. We found that the expression of SV2A was down-regulated in the hippocampus of AD patients, yet the relation of SV2A to other hallmarks of AD pathology such as amyloid precursor protein (APP), β-amyloid (Aβ), and Tau protein is not thoroughly clear. In addition, SV2A colocalized with APP and was down-regulated at Aβ deposition. Moreover, we found that SV2A deficiency leads to a simultaneous increase in Aβ and Tau hyperphosphorylation, while SV2A overexpression was associated with downregulation of β-site APP cleaving enzyme 1 and apolipoprotein E genes. In addition, evidence gained in the study points to the phosphatidylinositol 3-kinase signaling pathway as a possible mediator in SV2A regulation influencing the incidence and development of AD. With limited effective diagnostic methods for AD, a close interplay between SV2A and AD-related proteins demonstrated in our study may provide novel and innovative diagnostic and therapeutic opportunities.
Collapse
Affiliation(s)
- Yanyan Kong
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Lin Huang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Weihao Li
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Xuanting Liu
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Yinping Zhou
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Cuiping Liu
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Shibo Zhang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhengwei Zhang
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Donglang Jiang
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Weiyan Zhou
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Ruiqing Ni
- Institute for Biomedical Engineering, ETH Zurich, University of Zurich, Zurich, Switzerland
| | - Chencheng Zhang
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bomin Sun
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
73
|
Ando M, Nobukawa S, Kikuchi M, Takahashi T. Identification of Electroencephalogram Signals in Alzheimer's Disease by Multifractal and Multiscale Entropy Analysis. Front Neurosci 2021; 15:667614. [PMID: 34262427 PMCID: PMC8273283 DOI: 10.3389/fnins.2021.667614] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 06/01/2021] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is a progressive neurodegenerative disease that primarily develops in old age. In recent years, it has been reported that early diagnosis of AD and early intervention significantly delays disease progression. Hence, early diagnosis and intervention are emphasized. As a diagnostic index for AD patients, evaluating the complexity of the dependence of the electroencephalography (EEG) signal on the temporal scale of Alzheimer's disease (AD) patients is effective. Multiscale entropy analysis and multifractal analysis have been performed individually, and their usefulness as diagnostic indicators has been confirmed, but the complemental relationship between these analyses, which may enhance diagnostic accuracy, has not been investigated. We hypothesize that combining multiscale entropy and fractal analyses may add another dimension to understanding the alteration of EEG dynamics in AD. In this study, we performed both multiscale entropy and multifractal analyses on EEGs from AD patients and healthy subjects. We found that the classification accuracy was improved using both techniques. These findings suggest that the use of multiscale entropy analysis and multifractal analysis may lead to the development of AD diagnostic tools.
Collapse
Affiliation(s)
- Momo Ando
- Graduate School of Information and Computer Science, Chiba Institute of Technology, Narashino, Japan
| | - Sou Nobukawa
- Graduate School of Information and Computer Science, Chiba Institute of Technology, Narashino, Japan.,Department of Computer Science, Chiba Institute of Technology, Narashino, Japan
| | - Mitsuru Kikuchi
- Department of Psychiatry and Behavioral Science, Kanazawa University, Ishikawa, Japan.,Research Center for Child Mental Development, Kanazawa University, Ishikawa, Japan
| | - Tetsuya Takahashi
- Research Center for Child Mental Development, Kanazawa University, Ishikawa, Japan.,Department of Neuropsychiatry, University of Fukui, Fukui, Japan.,Uozu Shinkei Sanatorium, Uozu, Japan
| |
Collapse
|
74
|
Roy J, Tsui KC, Ng J, Fung ML, Lim LW. Regulation of Melatonin and Neurotransmission in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22136841. [PMID: 34202125 PMCID: PMC8268832 DOI: 10.3390/ijms22136841] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/20/2022] Open
Abstract
Alzheimer’s disease is a neurodegenerative disorder associated with age, and is characterized by pathological markers such as amyloid-beta plaques and neurofibrillary tangles. Symptoms of AD include cognitive impairments, anxiety and depression. It has also been shown that individuals with AD have impaired neurotransmission, which may result from the accumulation of amyloid plaques and neurofibrillary tangles. Preclinical studies showed that melatonin, a monoaminergic neurotransmitter released from the pineal gland, is able to ameliorate AD pathologies and restore cognitive impairments. Theoretically, inhibition of the pathological progression of AD by melatonin treatment should also restore the impaired neurotransmission. This review aims to explore the impact of AD on neurotransmission, and whether and how melatonin can enhance neurotransmission via improving AD pathology.
Collapse
|
75
|
Karimani A, Ramezani N, Afkhami Goli A, Nazem Shirazi MH, Nourani H, Jafari AM. Subchronic neurotoxicity of diazinon in albino mice: Impact of oxidative stress, AChE activity, and gene expression disturbances in the cerebral cortex and hippocampus on mood, spatial learning, and memory function. Toxicol Rep 2021; 8:1280-1288. [PMID: 34277358 PMCID: PMC8261896 DOI: 10.1016/j.toxrep.2021.06.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 06/06/2021] [Accepted: 06/14/2021] [Indexed: 02/03/2023] Open
Abstract
Diazinon (DZN) with prominent neurotoxic effects perturbs CNS function via multiple mechanisms. This investigation intends to explore mood, spatial learning, and memory dysfunction, acetylcholine esterase (AChE) activity, and neurodegeneration-related gene expression in the cortex and hippocampus regions of mice exposed to DZN for 63 consecutive days (subchronic exposure). Adult male albino mice were orally given sublethal DZN (DZNL = 0.1 mg/kg, DZNM = 1 mg/kg and DZNH = 10 mg/kg). All mice in the DZNH group died within 3 weeks postexposure. DZNL and DZNM caused body and brain weight loss (p < 0.05). Completing 9 weeks of DZN exposure, a marked decline in AChE activity and oxidative stress level was indicated in both brain regions (p < 0.05). Also, synaptophysin, vesicular acetylcholine transferase, and glutamate decarboxylase gene expressions were affected in both brain regions (p < 0.05). Furthermore, the present study revealed that DZN administration increased anxiety and depressive-like behaviors (p < 0.0001). Spatial learning and short- and long-memory were severely affected by DZNL and DZNM treatments (p < 0.0001). Taken together, subchronic exposure to low and medium doses of DZN can cause AChE inhibition, oxidative damage, and neurotransmitter disturbances in brain cells and induce neurodegeneration. These changes would impair mood, spatial learning, and memory function.
Collapse
Key Words
- AChE, acetylcholine esterase
- AD, Alzheimer’s disease
- Ach, acetylcholine
- COX-2, cyclooxygenase-2
- CX, cerebral cortex
- Cerebral cortex
- DZN, diazinon
- DZO, diazoxon
- Diazinon
- FRAP, ferric reducing antioxidant power
- FST, forced swim test
- GABA, ϒ-aminobutyric acid
- GAD65, glutamate decarboxylase 65
- HP, hippocampus
- Hippocampus
- LD50, lethal dose 50
- MB, marble burying test
- MDA, malondialdehyde
- MWM, Morris water maze test
- Memory
- NOAEL, no-observed-adverse-effect level
- Neurodegenerative diseases
- Ops, organophosphates
- PD, Parkinson’s disease
- RNS, reactive nitrogen species
- ROS, reactive oxygen species
- SEM, standard error of the mean
- SYP, synaptophysin
- Spatial learning
- VAChT, vesicular acetylcholine transferase
- qRT-PCR, quantitative reverse transcription-polymerase chain reaction
Collapse
Affiliation(s)
- Asieh Karimani
- Department of Toxicology, School of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Nasrin Ramezani
- Department of Toxicology, School of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Amir Afkhami Goli
- Department of Toxicology, School of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | - Hosein Nourani
- Department of Pathology, School of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Amir Moghaddam Jafari
- Department of Toxicology, School of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
76
|
Lee SH, Bolshakov VY, Shen J. Inactivation of Presenilin in inhibitory neurons results in decreased GABAergic responses and enhanced synaptic plasticity. Mol Brain 2021; 14:85. [PMID: 34034776 PMCID: PMC8152317 DOI: 10.1186/s13041-021-00796-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 05/18/2021] [Indexed: 11/10/2022] Open
Abstract
Mutations in the Presenilin genes are the major genetic cause of Alzheimer's disease (AD). Presenilin (PS) is highly expressed in the hippocampus, which is particularly vulnerable in AD. Previous studies of PS function in the hippocampus, however, focused exclusively on excitatory neurons. Whether PS regulates inhibitory neuronal function remained unknown. In the current study, we investigate PS function in GABAergic neurons by performing whole-cell and field-potential electrophysiological recordings using acute hippocampal slices from inhibitory neuron-specific PS conditional double knockout (IN-PS cDKO) mice at 2 months of age, before the onset of age-dependent loss of interneurons. We found that the frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) is reduced in hippocampal CA1 neurons of IN-PS cDKO mice, whereas the amplitude of sIPSCs is normal. Moreover, the efficacy of inhibitory neurotransmission as assessed with synaptic input/output relations for evoked mono- and di-synaptic IPSCs is markedly lowered in hippocampal CA1 neurons of IN-PS cDKO mice. Consistent with these findings, IN-PS cDKO mice display enhanced paired-pulse facilitation, frequency facilitation and long-term potentiation in the Schaffer collateral-CA1 pathway. Interestingly, depletion of intracellular Ca2+ stores by inhibition of sarcoendoplasmic reticulum Ca2+ ATPase results in a reduction of IPSC amplitude in control hippocampal neurons but not in IN-PS cDKO neurons, suggesting that impaired intracellular calcium homeostasis in the absence of PS may contribute to the deficiencies in inhibitory neurotransmission. Furthermore, the amplitude of IPSCs induced by short trains of presynaptic stimulation and paired-pulse ratio are decreased in IN-PS cDKO mice. These findings show that inactivation of PS in interneurons results in decreased GABAergic responses and enhanced synaptic plasticity in the hippocampus, providing additional evidence for the importance of PS in the regulation of synaptic plasticity and calcium homeostasis.
Collapse
Affiliation(s)
- Sang Hun Lee
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| | - Vadim Y Bolshakov
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
- Program in Neuroscience, Harvard Medical School, Boston, MA, 02115, USA
| | - Jie Shen
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Program in Neuroscience, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
77
|
Lauterborn JC, Scaduto P, Cox CD, Schulmann A, Lynch G, Gall CM, Keene CD, Limon A. Increased excitatory to inhibitory synaptic ratio in parietal cortex samples from individuals with Alzheimer's disease. Nat Commun 2021; 12:2603. [PMID: 33972518 PMCID: PMC8110554 DOI: 10.1038/s41467-021-22742-8] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 03/24/2021] [Indexed: 02/08/2023] Open
Abstract
Synaptic disturbances in excitatory to inhibitory (E/I) balance in forebrain circuits are thought to contribute to the progression of Alzheimer's disease (AD) and dementia, although direct evidence for such imbalance in humans is lacking. We assessed anatomical and electrophysiological synaptic E/I ratios in post-mortem parietal cortex samples from middle-aged individuals with AD (early-onset) or Down syndrome (DS) by fluorescence deconvolution tomography and microtransplantation of synaptic membranes. Both approaches revealed significantly elevated E/I ratios for AD, but not DS, versus controls. Gene expression studies in an independent AD cohort also demonstrated elevated E/I ratios in individuals with AD as compared to controls. These findings provide evidence of a marked pro-excitatory perturbation of synaptic E/I balance in AD parietal cortex, a region within the default mode network that is overly active in the disorder, and support the hypothesis that E/I imbalances disrupt cognition-related shifts in cortical activity which contribute to the intellectual decline in AD.
Collapse
Affiliation(s)
- Julie C Lauterborn
- Department of Anatomy and Neurobiology, University of California at Irvine, Irvine, CA, USA.
| | - Pietro Scaduto
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases. School of Medicine, University of Texas Medical Branch at Galveston, Galveston, USA
| | - Conor D Cox
- Department of Anatomy and Neurobiology, University of California at Irvine, Irvine, CA, USA
| | - Anton Schulmann
- National Institute of Mental Health, Human Genetics Branch, Bethesda, MD, USA
| | - Gary Lynch
- Department of Anatomy and Neurobiology, University of California at Irvine, Irvine, CA, USA
- Department of Psychiatry & Human Behavior, University of California at Irvine, Irvine, CA, 92697, USA
| | - Christine M Gall
- Department of Anatomy and Neurobiology, University of California at Irvine, Irvine, CA, USA
| | - C Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Agenor Limon
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases. School of Medicine, University of Texas Medical Branch at Galveston, Galveston, USA.
| |
Collapse
|
78
|
Artesunate restores the levels of inhibitory synapse proteins and reduces amyloid-β and C-terminal fragments (CTFs) of the amyloid precursor protein in an AD-mouse model. Mol Cell Neurosci 2021; 113:103624. [PMID: 33933588 DOI: 10.1016/j.mcn.2021.103624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease (AD) is the most frequent form of dementia, characterized histopathologically by the formation of amyloid plaques and neurofibrillary tangles in the brain. Amyloid β-peptide (Aβ) is a major component of amyloid plaques and is released together with carboxy-terminal fragments (CTFs) from the amyloid precursor protein (APP) through proteolytic cleavage, thought to contribute to synapse dysfunction and loss along the progression of AD. Artemisinins, primarily antimalarial drugs, reduce neuroinflammation and improve cognitive capabilities in mouse models of AD. Furthermore, artemisinins were demonstrated to target gephyrin, the main scaffold protein of inhibitory synapses and modulate GABAergic neurotransmission in vitro. Previously, we reported a robust decrease of inhibitory synapse proteins in the hippocampus of 12-month-old double transgenic APP-PS1 mice which overexpress in addition to the Swedish mutated form of the human APP a mutated presenilin 1 (PS1) gene and are characterized by a high plaque load at this age. Here, we provide in vivo evidence that treating these mice with artemisinin or its semisynthetic derivative artesunate in two different doses (10 mg/kg and 100 mg/kg), these compounds affect differently inhibitory synapse components, amyloid plaque load and APP-processing. Immunofluorescence microscopy demonstrated the rescue of gephyrin and γ2-GABAA-receptor protein levels in the brain of treated mice with both, artemisinin and artesunate, most efficiently with a low dose of artesunate. Remarkably, artemisinin reduced only in low dose the amyloid plaque load correlating with lower levels of mutated human APP (hAPPswe) whereas artesunate treatment in both doses resulted in significantly lower plaque numbers. Correspondingly, the level of APP-cleavage products, specifically the amount of CTFs in hippocampus homogenates was reduced significantly only by artesunate, in line with the findings in hAPPswe expressing cultured hippocampal neurons evidencing a concentration-dependent inhibition of CTF-release by artesunate already in the nanomolar range. Thus, our data support artemisinins as neuroprotective multi-target drugs, exhibiting a potent anti-amyloidogenic activity and reinforcing key proteins of inhibitory synapses.
Collapse
|
79
|
Du X, Shi Q, Zhao Y, Xie Y, Li X, Liu Q, Iqbal J, Zhang H, Liu X, Shen L. Se-Methylselenocysteine (SMC) Improves Cognitive Deficits by Attenuating Synaptic and Metabolic Abnormalities in Alzheimer's Mice Model: A Proteomic Study. ACS Chem Neurosci 2021; 12:1112-1132. [PMID: 33689275 DOI: 10.1021/acschemneuro.0c00549] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Se-methylselenocysteine (SMC) is a major selenocompound in selenium (Se) enriched plants and has been found to ameliorate neuropathology and cognitive deficits in triple-transgenic mice model of Alzheimer's disease (3 × Tg-AD mice). To explore the underlying molecular mechanisms, the present study is designed to elucidate the protein changes in the cortex of SMC-treated 3 × Tg-AD mice. After SMC supplementation, proteomic analysis revealed that 181, 271, and 41 proteins were identified as differentially expressed proteins (DEPs) between 3 × Tg-AD mice vs wild type (AD/WT group), SMC-treated AD mice vs AD (AD + SMC/AD), and AD + SMC/WT group, respectively. Among these, 138 proteins in the diseased group were reversed by SMC treatment. The DEPs in AD/WT group and AD + SMC/AD group were mainly related to metabolism, synapses, and antioxidant proteins, while their levels were decreased in AD mice but up-regulated after treating with SMC. In addition, we found reduced ATP levels and destroyed synaptic structures in the AD mice brains, which were significantly ameliorated upon SMC treatment. Our study suggests that energy metabolism disorders, abnormal amino acid metabolism, synaptic dysfunction, and oxidative stress may be the key pathogenic phenomena of AD. SMC reversed the expression of proteins associated with them, which might be the main mechanism of its intervention in AD.
Collapse
Affiliation(s)
- Xiubo Du
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, P. R. China
| | - Qingqing Shi
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, P. R. China
| | - Yuxi Zhao
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, P. R. China
| | - Yongli Xie
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, P. R. China
| | - Xuexia Li
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, P. R. China
| | - Qiong Liu
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, P. R. China
| | - Javed Iqbal
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, P. R. China
| | - Huajie Zhang
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, P. R. China
- Shenzhen Bay Laboratory, Shenzhen 518055, P.R. China
| | - Xukun Liu
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, P. R. China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, P. R. China
| | - Liming Shen
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Science and Oceanography, Shenzhen University, Shenzhen 518060, P. R. China
| |
Collapse
|
80
|
Borisova T, Pozdnyakova N, Dudarenko M, Krisanova N, Andronati S. GABAA receptor agonist cinazepam and its active metabolite 3-hydroxyphenazepam act differently at the presynaptic site. Eur Neuropsychopharmacol 2021; 45:39-51. [PMID: 33820715 DOI: 10.1016/j.euroneuro.2021.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 10/21/2022]
Abstract
Cinazepam C19H14BrClN2O5, ("LevanaⓇ ІC") a partial GABAA receptor agonist, and its active metabolite 3-hydroxyphenazepam C15H10BrClN2O2 were comparatively assessed in vitro using nerve terminals isolated from rat cortex (synaptosomes). At the presynaptic site, cinazepam (100 and 200 µM) facilitated synaptosomal transporter-mediated [3H]GABA uptake by enhancing both the initial rate and accumulation, and decreased the ambient level and transporter-mediated release of [3H]GABA. Whereas, 3-hydroxyphenazepam decreased the uptake and did not change the ambient synaptosomal level and transporter-mediated release of [3H]GABA. To exclude GABA transporter influence, NO-711, the transporter blocker, was applied and it was found that exocytotic release of [3H]GABA decreased, whereas tonic release of [3H]GABA was not changed in the presence of both cinazepam or 3-hydroxyphenazepam after treatment of synaptosomes with NO-711. In fluorimetric studies using potential- and pH-sensitive dyes rhodamine 6G and acridine orange, respectively, it was found that cinazepam hyperpolarized the synaptosomal plasma membrane, and increased synaptic vesicle acidification, whereas, 3-hydroxyphenazepam demonstrated opposite effects on these parameters. Therefore, action of cinazepam and its active metabolite 3-hydroxyphenazepam on GABAergic neurotransmission was different. Therapeutic effects of cinazepam can be associated with its ability to hyperpolarize the plasma membrane, to increase synaptic vesicle acidification and capacity of its active metabolite 3-hydroxyphenazepam to inhibit GABA transporter functioning.
Collapse
Affiliation(s)
- Tatiana Borisova
- The Department of Neurochemistry, The Palladin Institute of Biochemistry of the National Academy of Sciences of Ukraine, 9 Leontovicha Street, Kiev 01054, Ukraine.
| | - Natalia Pozdnyakova
- The Department of Neurochemistry, The Palladin Institute of Biochemistry of the National Academy of Sciences of Ukraine, 9 Leontovicha Street, Kiev 01054, Ukraine.
| | - Marina Dudarenko
- The Department of Neurochemistry, The Palladin Institute of Biochemistry of the National Academy of Sciences of Ukraine, 9 Leontovicha Street, Kiev 01054, Ukraine.
| | - Natalia Krisanova
- The Department of Neurochemistry, The Palladin Institute of Biochemistry of the National Academy of Sciences of Ukraine, 9 Leontovicha Street, Kiev 01054, Ukraine.
| | - Sergey Andronati
- The Department of Medicinal Chemistry, A.V. Bogatsky Physico-Chemical Institute of the National Academy of Sciences of Ukraine, 86 Lustdorfskaya doroga, 65080 Odessa, Ukraine.
| |
Collapse
|
81
|
Pasieka A, Panek D, Jończyk J, Godyń J, Szałaj N, Latacz G, Tabor J, Mezeiova E, Chantegreil F, Dias J, Knez D, Lu J, Pi R, Korabecny J, Brazzolotto X, Gobec S, Höfner G, Wanner K, Więckowska A, Malawska B. Discovery of multifunctional anti-Alzheimer's agents with a unique mechanism of action including inhibition of the enzyme butyrylcholinesterase and γ-aminobutyric acid transporters. Eur J Med Chem 2021; 218:113397. [PMID: 33838585 DOI: 10.1016/j.ejmech.2021.113397] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/31/2022]
Abstract
Looking for an effective anti-Alzheimer's agent is very challenging; however, a multifunctional ligand strategy may be a promising solution for the treatment of this complex disease. We herein present the design, synthesis and biological evaluation of novel hydroxyethylamine derivatives displaying unique, multiple properties that have not been previously reported. The original mechanism of action combines inhibitory activity against disease-modifying targets: β-secretase enzyme (BACE1) and amyloid β (Aβ) aggregation, along with an effect on targets associated with symptom relief - inhibition of butyrylcholinesterase (BuChE) and γ-aminobutyric acid transporters (GATs). Among the obtained molecules, compound 36 exhibited the most balanced and broad activity profile (eeAChE IC50 = 2.86 μM; eqBuChE IC50 = 60 nM; hBuChE IC50 = 20 nM; hBACE1 IC50 = 5.9 μM; inhibition of Aβ aggregation = 57.9% at 10 μM; mGAT1 IC50 = 10.96 μM; and mGAT2 IC50 = 19.05 μM). Moreover, we also identified 31 as the most potent mGAT4 and hGAT3 inhibitor (IC50 = 5.01 μM and IC50 = 2.95 μM, respectively), with high selectivity over other subtypes. Compounds 36 and 31 represent new anti-Alzheimer agents that can ameliorate cognitive decline and modify the progress of disease.
Collapse
Affiliation(s)
- Anna Pasieka
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Dawid Panek
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Jakub Jończyk
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Justyna Godyń
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Natalia Szałaj
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Gniewomir Latacz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Julia Tabor
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Eva Mezeiova
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic
| | - Fabien Chantegreil
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223, Brétigny sur Orge, France
| | - José Dias
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223, Brétigny sur Orge, France
| | - Damijan Knez
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Junfeng Lu
- Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Rongbiao Pi
- Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Jan Korabecny
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic
| | - Xavier Brazzolotto
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223, Brétigny sur Orge, France
| | - Stanislav Gobec
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Georg Höfner
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr, 5-13, 81377, Munich, Germany
| | - Klaus Wanner
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr, 5-13, 81377, Munich, Germany
| | - Anna Więckowska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Barbara Malawska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland.
| |
Collapse
|
82
|
Casani-Cubel J, Benlloch M, Sanchis-Sanchis CE, Marin R, Lajara-Romance JM, de la Rubia Orti JE. The Impact of Microbiota on the Pathogenesis of Amyotrophic Lateral Sclerosis and the Possible Benefits of Polyphenols. An Overview. Metabolites 2021; 11:120. [PMID: 33672485 PMCID: PMC7923408 DOI: 10.3390/metabo11020120] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/14/2021] [Accepted: 02/17/2021] [Indexed: 12/11/2022] Open
Abstract
The relationship between gut microbiota and neurodegenerative diseases is becoming clearer. Among said diseases amyotrophic lateral sclerosis (ALS) stands out due to its severity and, as with other chronic pathologies that cause neurodegeneration, gut microbiota could play a fundamental role in its pathogenesis. Therefore, polyphenols could be a therapeutic alternative due to their anti-inflammatory action and probiotic effect. Thus, the objective of our narrative review was to identify those bacteria that could have connection with the mentioned disease (ALS) and to analyze the benefits produced by administering polyphenols. Therefore, an extensive search was carried out selecting the most relevant articles published between 2005 and 2020 on the PubMed and EBSCO database on research carried out on cell, animal and human models of the disease. Thereby, after selecting, analyzing and debating the main articles on this topic, the bacteria related to the pathogenesis of ALS have been identified, among which we can positively highlight the presence mainly of Akkermansia muciniphila, but also Lactobacillus spp., Bifidobacterium spp. or Butyrivibrio fibrisolvens. Nevertheless, the presence of Escherichia coli or Ruminococcus torques stand out negatively for the disease. In addition, most of these bacteria are associated with molecular changes also linked to the pathogenesis of ALS. However, once the main polyphenols related to improvements in any of these three ALS models were assessed, many of them show positive results that could improve the prognosis of the disease. Nonetheless, epigallocatechin gallate (EGCG), curcumin and resveratrol are the polyphenols considered to show the most promising results as a therapeutic alternative for ALS through changes in microbiota.
Collapse
Affiliation(s)
- Julia Casani-Cubel
- Doctoral Degree School, Catholic University of Valencia San Vicente Mártir, 46001 Valencia, Spain;
| | - María Benlloch
- Department of Health Science, Catholic University San Vicente Mártir, 46001 Valencia, Spain;
| | | | - Raquel Marin
- Laboratory of Cellular Neurobiology, School of Medicine, Faculty of Health Sciences, University of La Laguna, 38190 Tenerife, Spain;
| | | | | |
Collapse
|
83
|
Ruan Z, Pathak D, Venkatesan Kalavai S, Yoshii-Kitahara A, Muraoka S, Bhatt N, Takamatsu-Yukawa K, Hu J, Wang Y, Hersh S, Ericsson M, Gorantla S, Gendelman HE, Kayed R, Ikezu S, Luebke JI, Ikezu T. Alzheimer's disease brain-derived extracellular vesicles spread tau pathology in interneurons. Brain 2021; 144:288-309. [PMID: 33246331 PMCID: PMC7880668 DOI: 10.1093/brain/awaa376] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/29/2020] [Accepted: 08/17/2020] [Indexed: 12/16/2022] Open
Abstract
Extracellular vesicles are highly transmissible and play critical roles in the propagation of tau pathology, although the underlying mechanism remains elusive. Here, for the first time, we comprehensively characterized the physicochemical structure and pathogenic function of human brain-derived extracellular vesicles isolated from Alzheimer's disease, prodromal Alzheimer's disease, and non-demented control cases. Alzheimer's disease extracellular vesicles were significantly enriched in epitope-specific tau oligomers in comparison to prodromal Alzheimer's disease or control extracellular vesicles as determined by dot blot and atomic force microscopy. Alzheimer's disease extracellular vesicles were more efficiently internalized by murine cortical neurons, as well as more efficient in transferring and misfolding tau, than prodromal Alzheimer's disease and control extracellular vesicles in vitro. Strikingly, the inoculation of Alzheimer's disease or prodromal Alzheimer's disease extracellular vesicles containing only 300 pg of tau into the outer molecular layer of the dentate gyrus of 18-month-old C57BL/6 mice resulted in the accumulation of abnormally phosphorylated tau throughout the hippocampus by 4.5 months, whereas inoculation of an equal amount of tau from control extracellular vesicles, isolated tau oligomers, or fibrils from the same Alzheimer's disease donor showed little tau pathology. Furthermore, Alzheimer's disease extracellular vesicles induced misfolding of endogenous tau in both oligomeric and sarkosyl-insoluble forms in the hippocampal region. Unexpectedly, phosphorylated tau was primarily accumulated in glutamic acid decarboxylase 67 (GAD67) GABAergic interneurons and, to a lesser extent, glutamate receptor 2/3-positive excitatory mossy cells, showing preferential extracellular vesicle-mediated GABAergic interneuronal tau propagation. Whole-cell patch clamp recordings of CA1 pyramidal cells showed significant reduction in the amplitude of spontaneous inhibitory post-synaptic currents. This was accompanied by reductions in c-fos+ GAD67+ neurons and GAD67+ neuronal puncta surrounding pyramidal neurons in the CA1 region, confirming reduced GABAergic transmission in this region. Our study posits a novel mechanism for the spread of tau in hippocampal GABAergic interneurons via brain-derived extracellular vesicles and their subsequent neuronal dysfunction.
Collapse
Affiliation(s)
- Zhi Ruan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Dhruba Pathak
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Srinidhi Venkatesan Kalavai
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Asuka Yoshii-Kitahara
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Satoshi Muraoka
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Nemil Bhatt
- Department of Neurology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Kayo Takamatsu-Yukawa
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jianqiao Hu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Yuzhi Wang
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Samuel Hersh
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Maria Ericsson
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neurosciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neurosciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Rakez Kayed
- Department of Neurology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Seiko Ikezu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jennifer I Luebke
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA 02118, USA
- Center for Systems Neuroscience, Boston University, Boston, MA 02118, USA
| | - Tsuneya Ikezu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
- Center for Systems Neuroscience, Boston University, Boston, MA 02118, USA
- Department of Neurology and Alzheimer’s Disease Center, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
84
|
Sears SM, Hewett SJ. Influence of glutamate and GABA transport on brain excitatory/inhibitory balance. Exp Biol Med (Maywood) 2021; 246:1069-1083. [PMID: 33554649 DOI: 10.1177/1535370221989263] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
An optimally functional brain requires both excitatory and inhibitory inputs that are regulated and balanced. A perturbation in the excitatory/inhibitory balance-as is the case in some neurological disorders/diseases (e.g. traumatic brain injury Alzheimer's disease, stroke, epilepsy and substance abuse) and disorders of development (e.g. schizophrenia, Rhett syndrome and autism spectrum disorder)-leads to dysfunctional signaling, which can result in impaired cognitive and motor function, if not frank neuronal injury. At the cellular level, transmission of glutamate and GABA, the principle excitatory and inhibitory neurotransmitters in the central nervous system control excitatory/inhibitory balance. Herein, we review the synthesis, release, and signaling of GABA and glutamate followed by a focused discussion on the importance of their transport systems to the maintenance of excitatory/inhibitory balance.
Collapse
Affiliation(s)
- Sheila Ms Sears
- Department of Biology, Program in Neuroscience, 2029Syracuse University, Syracuse, NY 13244, USA
| | - Sandra J Hewett
- Department of Biology, Program in Neuroscience, 2029Syracuse University, Syracuse, NY 13244, USA
| |
Collapse
|
85
|
Beato A, Gori A, Boucherle B, Peuchmaur M, Haudecoeur R. β-Carboline as a Privileged Scaffold for Multitarget Strategies in Alzheimer's Disease Therapy. J Med Chem 2021; 64:1392-1422. [PMID: 33528252 DOI: 10.1021/acs.jmedchem.0c01887] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The natural β-carboline alkaloids display similarities with neurotransmitters that can be favorably exploited to design bioactive and bioavailable drugs for Alzheimer's disease (AD) therapy. Several AD targets are currently and intensively being investigated, divided in different hypotheses: mainly the cholinergic, the amyloid β (Aβ), and the Tau hypotheses. To date, only symptomatic treatments are available involving acetylcholinesterase and NMDA inhibitors. On the basis of plethoric single-target structure-activity relationship studies, the β-carboline scaffold was identified as a powerful tool for fostering activity and molecular interactions with a wide range of AD-related targets. This knowledge can undoubtedly be used to design multitarget-directed ligands, a highly relevant strategy preferred in the context of multifactorial pathology with intricate etiology such as AD. In this review, we first individually discuss the AD targets of the β-carbolines, and then we focus on the multitarget strategies dedicated to the deliberate design of new efficient scaffolds.
Collapse
Affiliation(s)
| | - Anthonin Gori
- Univ. Grenoble Alpes, CNRS, DPM, 38000 Grenoble, France.,CHANEL Parfums Beauté, F-93500 Pantin, France
| | | | | | | |
Collapse
|
86
|
Mohammadnejad A, Soerensen M, Baumbach J, Mengel‐From J, Li W, Lund J, Li S, Christiansen L, Christensen K, Hjelmborg JVB, Tan Q. Novel DNA methylation marker discovery by assumption-free genome-wide association analysis of cognitive function in twins. Aging Cell 2021; 20:e13293. [PMID: 33528912 PMCID: PMC7884045 DOI: 10.1111/acel.13293] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 11/23/2020] [Accepted: 12/01/2020] [Indexed: 01/23/2023] Open
Abstract
Privileged by rapid increase in available epigenomic data, epigenome-wide association studies (EWAS) are to make a profound contribution to understand the molecular mechanism of DNA methylation in cognitive aging. Current statistical methods used in EWAS are dominated by models based on multiple assumptions, for example, linear relationship between molecular profiles and phenotype, normal distribution for the methylation data and phenotype. In this study, we applied an assumption-free method, the generalized correlation coefficient (GCC), and compare it to linear models, namely the linear mixed model and kinship model. We use DNA methylation associated with a cognitive score in 400 and 206 twins as discovery and replication samples respectively. DNA methylation associated with cognitive function using GCC, linear mixed model, and kinship model, identified 65 CpGs (p < 1e-04) from discovery sample displaying both nonlinear and linear correlations. Replication analysis successfully replicated 9 of these top CpGs. When combining results of GCC and linear models to cover diverse patterns of relationships, we identified genes like KLHDC4, PAPSS2, and MRPS18B as well as pathways including focal adhesion, axon guidance, and some neurological signaling. Genomic region-based analysis found 15 methylated regions harboring 11 genes, with three verified in gene expression analysis, also the 11 genes were related to top functional clusters including neurohypophyseal hormone and maternal aggressive behaviors. The GCC approach detects valuable methylation sites missed by traditional linear models. A combination of methylation markers from GCC and linear models enriched biological pathways sensible in neurological function that could implicate cognitive performance and cognitive aging.
Collapse
Affiliation(s)
- Afsaneh Mohammadnejad
- Unit of Epidemiology, Biostatistics and BiodemographyDepartment of Public HealthUniversity of Southern DenmarkOdenseDenmark
| | - Mette Soerensen
- Unit of Epidemiology, Biostatistics and BiodemographyDepartment of Public HealthUniversity of Southern DenmarkOdenseDenmark
- Unit of Human GeneticsDepartment of Clinical ResearchUniversity of Southern DenmarkOdenseDenmark
- Department of Clinical Biochemistry and PharmacologyOdense University HospitalOdenseDenmark
| | - Jan Baumbach
- Computational BiomedicineDepartment of Mathematics and Computer ScienceUniversity of Southern DenmarkOdenseDenmark
- Chair of Experimental BioinformaticsTUM School of Life SciencesTechnical University of MunichMunichGermany
| | - Jonas Mengel‐From
- Unit of Epidemiology, Biostatistics and BiodemographyDepartment of Public HealthUniversity of Southern DenmarkOdenseDenmark
- Unit of Human GeneticsDepartment of Clinical ResearchUniversity of Southern DenmarkOdenseDenmark
| | - Weilong Li
- Unit of Epidemiology, Biostatistics and BiodemographyDepartment of Public HealthUniversity of Southern DenmarkOdenseDenmark
- Population Research UnitFaculty of Social SciencesUniversity of HelsinkiHelsinkiFinland
| | - Jesper Lund
- Unit of Epidemiology, Biostatistics and BiodemographyDepartment of Public HealthUniversity of Southern DenmarkOdenseDenmark
- Digital Health & Machine Learning Research GroupHasso Plattner Institute for Digital EngineeringPotsdamGermany
| | - Shuxia Li
- Unit of Epidemiology, Biostatistics and BiodemographyDepartment of Public HealthUniversity of Southern DenmarkOdenseDenmark
| | - Lene Christiansen
- Unit of Epidemiology, Biostatistics and BiodemographyDepartment of Public HealthUniversity of Southern DenmarkOdenseDenmark
- Department of Clinical ImmunologyCopenhagen University HospitalRigshospitaletCopenhagen ØDenmark
| | - Kaare Christensen
- Unit of Epidemiology, Biostatistics and BiodemographyDepartment of Public HealthUniversity of Southern DenmarkOdenseDenmark
- Unit of Human GeneticsDepartment of Clinical ResearchUniversity of Southern DenmarkOdenseDenmark
| | - Jacob V. B. Hjelmborg
- Unit of Epidemiology, Biostatistics and BiodemographyDepartment of Public HealthUniversity of Southern DenmarkOdenseDenmark
| | - Qihua Tan
- Unit of Epidemiology, Biostatistics and BiodemographyDepartment of Public HealthUniversity of Southern DenmarkOdenseDenmark
- Unit of Human GeneticsDepartment of Clinical ResearchUniversity of Southern DenmarkOdenseDenmark
| |
Collapse
|
87
|
Zheng Q, Bi R, Xu M, Zhang DF, Tan LW, Lu YP, Yao YG. Exploring the Genetic Association of the ABAT Gene with Alzheimer's Disease. Mol Neurobiol 2021; 58:1894-1903. [PMID: 33404980 DOI: 10.1007/s12035-020-02271-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022]
Abstract
Accumulating evidence demonstrated that GABAergic dysfunction contributes to the pathogenesis of Alzheimer's disease (AD). The GABA aminotransferase (ABAT) gene encodes a mitochondrial GABA transaminase and plays key roles in the biogenesis and metabolism of gamma-aminobutyric acid (GABA), which is a major inhibitory neurotransmitter. In this study, we performed an integrative study at the genetic and expression levels to investigate the potential genetic association between the ABAT gene and AD. Through re-analyzing data from the currently largest meta-analysis of AD genome-wide association study (GWAS), we identified genetic variants in the 3'-UTR of ABAT as the top AD-associated SNPs (P < 1 × 10-4) in this gene. Functional annotation of these AD-associated SNPs indicated that these SNPs are located in the regulatory regions of transcription factors or/and microRNAs. Expression quantitative trait loci (eQTL) analysis and luciferase reporter assay showed that the AD risk alleles of these SNPs were associated with a reduced expression level of ABAT. Further analysis of mRNA expression data and single-cell transcriptome data of AD patients showed that ABAT reduction in the neuron is an early event during AD development. Overall, our results indicated that ABAT genetic variants may be associated with AD through affecting its mRNA expression. An abnormal level of ABAT will lead to a disturbance of the GABAergic signal pathway in AD brains.
Collapse
Affiliation(s)
- Quanzhen Zheng
- College of Life Sciences, Anhui Normal University, Wuhu, 241002, Anhui, China.,Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China
| | - Rui Bi
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Min Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China
| | - Deng-Feng Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China
| | - Li-Wen Tan
- Mental Health Institute of the Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Ya-Ping Lu
- College of Life Sciences, Anhui Normal University, Wuhu, 241002, Anhui, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China. .,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China. .,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
88
|
Kwakowsky A, Waldvogel HJ, Faull RLM. Therapeutic potential of alpha 5 subunit containing GABA A receptors in Alzheimer's disease. Neural Regen Res 2021; 16:1550-1551. [PMID: 33433477 PMCID: PMC8323681 DOI: 10.4103/1673-5374.300987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
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
| | - 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 M Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
89
|
Stolero N, Frenkel D. The dialog between neurons and microglia in Alzheimer's disease: The neurotransmitters view. J Neurochem 2020; 158:1412-1424. [PMID: 33314073 DOI: 10.1111/jnc.15262] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 11/12/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023]
Abstract
Microglia play a vital role in maintaining brain homeostasis. Their continuous sensing of surrounding micro-environments is crucial for their activity. Cross talk between specific neurons and microglia might occur through specific neurotransmitter receptors on microglia. Impairment with this interaction might result in pathological activity of microglia against potential insults. The reason for this activity in many neurodegenerative diseases such as Alzheimer's disease (AD) is not known. However, several papers report of the effects of different neurotransmitter agonists on microglial cells function that relate to their activity in AD. This review aims to summarize those works and to raise potential fundamental questions for future research.
Collapse
Affiliation(s)
- Nofar Stolero
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Dan Frenkel
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
90
|
Mohammadnejad A, Nygaard M, Li S, Zhang D, Xu C, Li W, Lund J, Christiansen L, Baumbach J, Christensen K, Hjelmborg JVB, Tan Q. Generalized correlation coefficient for genome-wide association analysis of cognitive ability in twins. Aging (Albany NY) 2020; 12:22457-22494. [PMID: 33232274 PMCID: PMC7746382 DOI: 10.18632/aging.104198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/29/2020] [Indexed: 12/19/2022]
Abstract
Despite a strong genetic background in cognitive function only a limited number of single nucleotide polymorphisms (SNPs) have been found in genome-wide association studies (GWASs). We hypothesize that this is partially due to mis-specified modeling concerning phenotype distribution as well as the relationship between SNP dosage and the level of the phenotype. To overcome these issues, we introduced an assumption-free method based on generalized correlation coefficient (GCC) in a GWAS of cognitive function in Danish and Chinese twins to compare its performance with traditional linear models. The GCC-based GWAS identified two significant SNPs in Danish samples (rs71419535, p = 1.47e-08; rs905838, p = 1.69e-08) and two significant SNPs in Chinese samples (rs2292999, p = 9.27e-10; rs17019635, p = 2.50e-09). In contrast, linear models failed to detect any genome-wide significant SNPs. The number of top significant genes overlapping between the two samples in the GCC-based GWAS was higher than when applying linear models. The GCC model identified significant genetic variants missed by conventional linear models, with more replicated genes and biological pathways related to cognitive function. Moreover, the GCC-based GWAS was robust in handling correlated samples like twin pairs. GCC is a useful statistical method for GWAS that complements traditional linear models for capturing genetic effects beyond the additive assumption.
Collapse
Affiliation(s)
- Afsaneh Mohammadnejad
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Marianne Nygaard
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Shuxia Li
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao, China
| | - Chunsheng Xu
- Qingdao Center for Disease Control and Prevention, Qingdao, China
| | - Weilong Li
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Jesper Lund
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Lene Christiansen
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen Ø, Denmark
| | - Jan Baumbach
- Computational Biomedicine, Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
- Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Kaare Christensen
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
- Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jacob v. B. Hjelmborg
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Qihua Tan
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
- Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
91
|
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
|
92
|
Zhao Y, Chen H, Iqbal J, Liu X, Zhang H, Xiao S, Jin N, Yao F, Shen L. Targeted metabolomics study of early pathological features in hippocampus of triple transgenic Alzheimer's disease male mice. J Neurosci Res 2020; 99:927-946. [PMID: 33197957 DOI: 10.1002/jnr.24750] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a serious neurodegenerative disease in people of age 65 or above. The detailed etiology and pathogenesis of AD have not been elucidated yet. In this study, the hippocampi of 2- and 6-month-old triple transgenic Alzheimer's disease male mice and age-sex-matched wild-type (WT) mice were analyzed by using targeted metabolomics approach. Compared with WT mice, 24 and 60 metabolites were found with significant differences in 2- and 6-month-old AD mice. Among these, 14 metabolites were found common while 10 metabolites showed consistent variable trends in both groups. These differential metabolites are found associated with amino acid, lipid, vitamin, nucleotide-related base, neurotransmitter and energy metabolisms, and oxidative stress. The results suggest that these differential metabolites might play a critical role in AD pathophysiology, and may serve as potential biomarkers for AD. Moreover, the results highlight the involvement of abnormal purine, pyrimidine, arginine, and proline metabolism, along with glycerophospholipid metabolism in early pathology of AD. For the first time, several differential metabolites are found to be associated with AD in this study. Targeted metabolomics can be used for rapid and accurate quantitative analysis of specific target metabolites associated with AD.
Collapse
Affiliation(s)
- Yuxi Zhao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| | - Haiquan Chen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| | - Javed Iqbal
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| | - Xukun Liu
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| | - Huajie Zhang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China.,Shenzhen Bay Laboratory, Shenzhen, P.R. China
| | - Shifeng Xiao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| | - Na Jin
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| | - Fang Yao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China.,Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Shenzhen, P.R. China
| | - Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| |
Collapse
|
93
|
Calvo-Flores Guzmán B, Elizabeth Chaffey T, Hansika Palpagama T, Waters S, Boix J, Tate WP, Peppercorn K, Dragunow M, Waldvogel HJ, Faull RLM, Kwakowsky A. The Interplay Between Beta-Amyloid 1-42 (Aβ 1-42)-Induced Hippocampal Inflammatory Response, p-tau, Vascular Pathology, and Their Synergistic Contributions to Neuronal Death and Behavioral Deficits. Front Mol Neurosci 2020; 13:522073. [PMID: 33224025 PMCID: PMC7667153 DOI: 10.3389/fnmol.2020.552073] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD), the most common chronic neurodegenerative disorder, has complex neuropathology. The principal neuropathological hallmarks of the disease are the deposition of extracellular β-amyloid (Aβ) plaques and neurofibrillary tangles (NFTs) comprised of hyperphosphorylated tau (p-tau) protein. These changes occur with neuroinflammation, a compromised blood-brain barrier (BBB) integrity, and neuronal synaptic dysfunction, all of which ultimately lead to neuronal cell loss and cognitive deficits in AD. Aβ1-42 was stereotaxically administered bilaterally into the CA1 region of the hippocampi of 18-month-old male C57BL/6 mice. This study aimed to characterize, utilizing immunohistochemistry and behavioral testing, the spatial and temporal effects of Aβ1-42 on a broad set of parameters characteristic of AD: p-tau, neuroinflammation, vascular pathology, pyramidal cell survival, and behavior. Three days after Aβ1-42 injection and before significant neuronal cell loss was detected, acute neuroinflammatory and vascular responses were observed. These responses included the up-regulation of glial fibrillary acidic protein (GFAP), cell adhesion molecule-1 (PECAM-1, also known as CD31), fibrinogen labeling, and an increased number of activated astrocytes and microglia in the CA1 region of the hippocampus. From day 7, there was significant pyramidal cell loss in the CA1 region of the hippocampus, and by 30 days, significant localized up-regulation of p-tau, GFAP, Iba-1, CD31, and alpha-smooth muscle actin (α-SMA) in the Aβ1-42-injected mice compared with controls. These molecular changes in Aβ1-42-injected mice were accompanied by cognitive deterioration, as demonstrated by long-term spatial memory impairment. This study is reporting a comprehensive examination of a complex set of parameters associated with intrahippocampal administration of Aβ1-42 in mice, their spatiotemporal interactions and combined contribution to the disease progression. We show that a single Aβ injection can reproduce aspects of the inflammatory, vascular, and p-tau induced pathology occurring in the AD human brain that lead to cognitive deficits.
Collapse
Affiliation(s)
- 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
| | - Tessa Elizabeth Chaffey
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Thulani Hansika Palpagama
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Sarah Waters
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Jordi Boix
- Centre for Brain Research, NeuroDiscovery Behavioural Unit, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Warren Perry Tate
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Katie Peppercorn
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Michael Dragunow
- Centre for Brain Research, Department of Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Henry John Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard Lewis Maxwell Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
94
|
Piras IS, Krate J, Delvaux E, Nolz J, Mastroeni DF, Persico AM, Jepsen WM, Beach TG, Huentelman MJ, Coleman PD. Transcriptome Changes in the Alzheimer's Disease Middle Temporal Gyrus: Importance of RNA Metabolism and Mitochondria-Associated Membrane Genes. J Alzheimers Dis 2020; 70:691-713. [PMID: 31256118 DOI: 10.3233/jad-181113] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We used Illumina Human HT-12 v4 arrays to compare RNA expression of middle temporal gyrus (MTG; BA21) in Alzheimer's disease (AD = 97) and non-demented controls (ND = 98). A total of 938 transcripts were highly differentially expressed (adj p < 0.01; log2 FC ≥ |0.500|, with 411 overexpressed and 527 underexpressed in AD. Our results correlated with expression profiling in neurons from AD and ND obtained by laser capture microscopy in MTG from an independent dataset (log2 FC correlation: r = 0.504; p = 2.2e-16). Additionally, selected effects were validated by qPCR. ANOVA analysis yielded no difference between genders in response to AD, but some gender specific genes were detected (e.g., IL8 and AGRN in males, and HSPH1 and GRM1 in females). Several transcripts were associated with Braak staging (e.g., AEBP1 and DNALI1), antemortem MMSE (e.g., AEBP1 and GFAP), and tangle density (e.g., RNU1G2, and DNALI1). At the pathway level, we detected enrichment of synaptic vesicle processes and GABAergic transmission genes. Finally, applying the Weighted Correlation Network Analysis, we identified four expression modules enriched for neuronal and synaptic genes, mitochondria-associated membrane, chemical stimulus and olfactory receptor and non-coding RNA metabolism genes. Our results represent an extensive description of MTG mRNA profiling in a large sample of AD and ND. These data provide a list of genes associated with AD, and correlated to neurofibrillary tangles density. In addition, these data emphasize the importance of mitochondrial membranes and transcripts related to olfactory receptors in AD.
Collapse
Affiliation(s)
- Ignazio S Piras
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Jonida Krate
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Elaine Delvaux
- Biodesign Institute, Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, USA
| | - Jennifer Nolz
- Biodesign Institute, Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, USA
| | - Diego F Mastroeni
- Biodesign Institute, Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, USA
| | - Antonio M Persico
- Unit of Child and Adolescent Neuropsychiatry, "Gaetano Martino" University Hospital, University of Messina, Messina, Italy.,Mafalda Luce Center for Pervasive Developmental Disorders, Milan, Italy
| | - Wayne M Jepsen
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Thomas G Beach
- Civin Laboratory of Neuropathology at Banner Sun Health Research Institute, Sun City, AZ, US
| | - Matthew J Huentelman
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Paul D Coleman
- Biodesign Institute, Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
95
|
Zhuang Z, Yang R, Wang W, Qi L, Huang T. Associations between gut microbiota and Alzheimer's disease, major depressive disorder, and schizophrenia. J Neuroinflammation 2020; 17:288. [PMID: 33008395 PMCID: PMC7532639 DOI: 10.1186/s12974-020-01961-8] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/23/2020] [Indexed: 12/11/2022] Open
Abstract
Background Growing evidence has shown that alterations in the gut microbiota composition were associated with a variety of neuropsychiatric conditions. However, whether such associations reflect causality remains unknown. We aimed to reveal the causal relationships among gut microbiota, metabolites, and neuropsychiatric disorders including Alzheimer’s disease (AD), major depressive disorder (MDD), and schizophrenia (SCZ). Methods A two-sample bi-directional Mendelian randomization analysis was performed by using genetic variants from genome-wide association studies as instrumental variables for gut microbiota, metabolites, AD, MDD, and SCZ, respectively. Results We found suggestive associations of host-genetic-driven increase in Blautia (OR, 0.88; 95%CI, 0.79–0.99; P = 0.028) and elevated γ-aminobutyric acid (GABA) (0.96; 0.92–1.00; P = 0.034), a downstream product of Blautia-dependent arginine metabolism, with a lower risk of AD. Genetically increased Enterobacteriaceae family and Enterobacteriales order were potentially associated with a higher risk of SCZ (1.09; 1.00–1.18; P = 0.048), while Gammaproteobacteria class (0.90; 0.83–0.98; P = 0.011) was related to a lower risk for SCZ. Gut production of serotonin was potentially associated with an increased risk of SCZ (1.07; 1.00–1.15; P = 0.047). Furthermore, genetically increased Bacilli class was related to a higher risk of MDD (1.07; 1.02–1.12; P = 0.010). In the other direction, neuropsychiatric disorders altered gut microbiota composition. Conclusions These data for the first time provide evidence of potential causal links between gut microbiome and AD, MDD, and SCZ. GABA and serotonin may play an important role in gut microbiota-host crosstalk in AD and SCZ, respectively. Further investigations in understanding the underlying mechanisms of associations between gut microbiota and AD, MDD, and SCZ are required.
Collapse
Affiliation(s)
- Zhenhuang Zhuang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, 38 Xueyuan Road, Beijing, 100191, China
| | - Ruotong Yang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, 38 Xueyuan Road, Beijing, 100191, China
| | - Wenxiu Wang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, 38 Xueyuan Road, Beijing, 100191, China
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA. .,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Tao Huang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, 38 Xueyuan Road, Beijing, 100191, China. .,Department of Global Health, School of Public Health, Peking University, Beijing, 100191, China. .,Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing, 100191, China. .,Center for Intelligent Public Health, Institute for Artificial Intelligence, Peking University, Beijing, 100191, China.
| |
Collapse
|
96
|
Ciminelli BM, Menduti G, Benussi L, Ghidoni R, Binetti G, Squitti R, Rongioletti M, Nica S, Novelletto A, Rossi L, Malaspina P. Polymorphic Genetic Markers of the GABA Catabolism Pathway in Alzheimer's Disease. J Alzheimers Dis 2020; 77:301-311. [PMID: 32804142 DOI: 10.3233/jad-200429] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND The compilation of a list of genetic modifiers in Alzheimer's disease (AD) is an open research field. The GABAergic system is affected in several neurological disorders but its role in AD is largely understudied. OBJECTIVE/METHODS As an explorative study, we considered variants in genes of GABA catabolism (ABAT, ALDH5A1, AKR7A2), and APOE in 300 Italian patients and 299 controls. We introduce a recent multivariate method to take into account the individual APOE genotype, thus controlling for the effect of the discrepant allele distributions in cases versus controls. We add a genotype-phenotype analysis based on age at onset and the Mini-Mental State Evaluation score. RESULTS On the background of strongly divergent APOE allele distributions in AD versus controls, two genotypic interactions that represented a subtle but significant peculiarity of the AD cohort emerged. The first is between ABAT and APOE, and the second between some ALDH5A1 genotypes and APOE. Decreased SSADH activity is predicted in AD carriers of APOEɛ4, representing an additional suggestion for increased oxidative damage. CONCLUSION We identified a difference between AD and controls, not in a shift of the allele frequencies at genes of the GABA catabolism pathway, but rather in gene interactions peculiar of the AD cohort. The emerging view is that of a multifactorial contribution to the disease, with a main risk factor (APOE), and additional contributions by the variants here considered. We consider genes of the GABA degradation pathway good candidates as modifiers of AD, contributing to energy impairment in AD brain.
Collapse
Affiliation(s)
| | | | - Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Giuliano Binetti
- MAC Memory Clinic and Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Rosanna Squitti
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Mauro Rongioletti
- Department of Laboratory Medicine, Research and Development Division, Fatebenefratelli Hospital, Isola Tiberina, Rome, Italy
| | - Sabrina Nica
- Department of Biology, University of Rome Tor Vergata, Italy
| | | | - Luisa Rossi
- Department of Biology, University of Rome Tor Vergata, Italy
| | | |
Collapse
|
97
|
Xu Y, Zhao M, Han Y, Zhang H. GABAergic Inhibitory Interneuron Deficits in Alzheimer's Disease: Implications for Treatment. Front Neurosci 2020; 14:660. [PMID: 32714136 PMCID: PMC7344222 DOI: 10.3389/fnins.2020.00660] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/28/2020] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized clinically by severe cognitive deficits and pathologically by amyloid plaques, neuronal loss, and neurofibrillary tangles. Abnormal amyloid β-protein (Aβ) deposition in the brain is often thought of as a major initiating factor in AD neuropathology. However, gamma-aminobutyric acid (GABA) inhibitory interneurons are resistant to Aβ deposition, and Aβ decreases synaptic glutamatergic transmission to decrease neural network activity. Furthermore, there is now evidence suggesting that neural network activity is aberrantly increased in AD patients and animal models due to functional deficits in and decreased activity of GABA inhibitory interneurons, contributing to cognitive deficits. Here we describe the roles played by excitatory neurons and GABA inhibitory interneurons in Aβ-induced cognitive deficits and how altered GABA interneurons regulate AD neuropathology. We also comprehensively review recent studies on how GABA interneurons and GABA receptors can be exploited for therapeutic benefit. GABA interneurons are an emerging therapeutic target in AD, with further clinical trials urgently warranted.
Collapse
Affiliation(s)
- Yilan Xu
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| | - Manna Zhao
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| | - Yuying Han
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| | - Heng Zhang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| |
Collapse
|
98
|
Menduti G, Vitaliti A, Capo CR, Lettieri-Barbato D, Aquilano K, Malaspina P, Rossi L. SSADH Variants Increase Susceptibility of U87 Cells to Mitochondrial Pro-Oxidant Insult. Int J Mol Sci 2020; 21:ijms21124374. [PMID: 32575506 PMCID: PMC7352567 DOI: 10.3390/ijms21124374] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 11/16/2022] Open
Abstract
Succinate semialdehyde dehydrogenase (SSADH) is a mitochondrial enzyme, encoded by ALDH5A1, mainly involved in γ-aminobutyric acid (GABA) catabolism and energy supply of neuronal cells, possibly contributing to antioxidant defense. This study aimed to further investigate the antioxidant role of SSADH, and to verify if common SNPs of ALDH5A1 may affect SSADH activity, stability, and mitochondrial function. In this study, we used U87 glioblastoma cells as they represent a glial cell line. These cells were transiently transfected with a cDNA construct simultaneously harboring three SNPs encoding for a triple mutant (TM) SSADH protein (p.G36R/p.H180Y/p.P182L) or with wild type (WT) cDNA. SSADH activity and protein level were measured. Cell viability, lipid peroxidation, mitochondrial morphology, membrane potential (ΔΨ), and protein markers of mitochondrial stress were evaluated upon Paraquat treatment, in TM and WT transfected cells. TM transfected cells show lower SSADH protein content and activity, fragmented mitochondria, higher levels of peroxidized lipids, and altered ΔΨ than WT transfected cells. Upon Paraquat treatment, TM cells show higher cell death, lipid peroxidation, 4-HNE protein adducts, and lower ΔΨ, than WT transfected cells. These results reinforce the hypothesis that SSADH contributes to cellular antioxidant defense; furthermore, common SNPs may produce unstable, less active SSADH, which could per se negatively affect mitochondrial function and, under oxidative stress conditions, fail to protect mitochondria.
Collapse
Affiliation(s)
- Giovanna Menduti
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; (G.M.); (A.V.); (C.R.C.); (D.L.-B.); (K.A.); (P.M.)
| | - Alessandra Vitaliti
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; (G.M.); (A.V.); (C.R.C.); (D.L.-B.); (K.A.); (P.M.)
| | - Concetta Rosa Capo
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; (G.M.); (A.V.); (C.R.C.); (D.L.-B.); (K.A.); (P.M.)
| | - Daniele Lettieri-Barbato
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; (G.M.); (A.V.); (C.R.C.); (D.L.-B.); (K.A.); (P.M.)
- IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; (G.M.); (A.V.); (C.R.C.); (D.L.-B.); (K.A.); (P.M.)
| | - Patrizia Malaspina
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; (G.M.); (A.V.); (C.R.C.); (D.L.-B.); (K.A.); (P.M.)
| | - Luisa Rossi
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; (G.M.); (A.V.); (C.R.C.); (D.L.-B.); (K.A.); (P.M.)
- Correspondence:
| |
Collapse
|
99
|
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: 111] [Impact Index Per Article: 22.2] [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
|
100
|
Neurotransmitter imbalance dysregulates brain dynamic fluidity in frontotemporal degeneration. Neurobiol Aging 2020; 94:176-184. [PMID: 32629312 DOI: 10.1016/j.neurobiolaging.2020.05.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/19/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022]
Abstract
Frontotemporal degeneration (FTD) is characterized by reduced global brain flexibility along with GABAergic/glutamatergic neurotransmitter deficits. We aimed to assess the relationship between dynamical properties of time-varying whole-brain network connectivity as well as static large-scale networks and neurotransmitter imbalance using resting-state functional MRI and transcranial magnetic stimulation (TMS) in sixty-six patients with FTD. We assessed GABAergic and glutamatergic neurotransmission by TMS, considering short- and long-interval intracortical inhibition and intracortical facilitation, and large-scale networks connectivity as well as four indexes of meta-state dynamic fluidity: (1) number of distinct meta-states, (2) number of switches from one meta-state to another, (3) span of the realized meta-states, and (4) total distance traveled in the state space. No significant correlations between TMS parameters and large-scale networks connectivity were observed. However, we observed a significant correlation between short-interval intracortical inhibition-intracortical facilitation and four meta-states (all indexes p < 0.02, false discovery rate-corrected). This study suggests that neurotransmitter imbalance dysregulates brain dynamic fluidity, linking microscopic and macroscopic changes in FTD.
Collapse
|