1
|
Xu C, Liu J, Guo M, Wang J, Bai X, Zhang C, Luan X, Pei H, Liu H, Lv X, Ye X, Tang B, Chen M. Targeting ApoE-KCC2 Signaling Rescues GABAergic Synaptic Dysfunction and Depression-like Behaviors in Mice. RESEARCH (WASHINGTON, D.C.) 2025; 8:0746. [PMID: 40530388 PMCID: PMC12173456 DOI: 10.34133/research.0746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 05/27/2025] [Accepted: 05/28/2025] [Indexed: 06/20/2025]
Abstract
Apolipoprotein E (ApoE) has been implicated in neurodegenerative diseases; however, its function and underlying mechanisms in depression remain elusive. In this study, we employed chronic social defeat stress (CSDS) to establish a mouse model of depression and observed significantly reduced ApoE expression in the hippocampus. By leveraging ApoE knockout (ApoE-/- ) and knockdown (ApoE-KD) mouse models, we demonstrated that ApoE deficiency induced depression-like behaviors, which were closely associated with impaired GABAergic synaptic transmission and down-regulation of ApoE receptors and K+-Cl- cotransporter 2 (KCC2). In addition, we found an interaction between KCC2 and the ApoE receptor low-density lipoprotein receptor (LDLR) through coimmunoprecipitation analysis. Moreover, overexpression of ApoE or targeted activation of GABAergic neurons in the hippocampus significantly reversed depression-like behaviors in both CSDS-exposed and ApoE-KD mice. Lastly, treatment with KCC2 activators, CLP290 and CLP257, restored the expression levels of KCC2 and the GABAAR α1 subunit, significantly alleviating depression-like behaviors induced by CSDS or ApoE-KD. Together, our results elucidate the pivotal role of ApoE in the pathophysiology of depression and highlight the ApoE-KCC2 signaling pathway as a potential target for developing innovative antidepressant therapies.
Collapse
Affiliation(s)
- Chengyuan Xu
- Department of Pharmacology, School of Pharmaceutical Sciences,
Anhui Medical University, Hefei 230032, Anhui, China
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital,
Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Jing Liu
- Department of Pharmacology, School of Pharmaceutical Sciences,
Anhui Medical University, Hefei 230032, Anhui, China
| | - Mengru Guo
- Department of Pharmacology, School of Pharmaceutical Sciences,
Anhui Medical University, Hefei 230032, Anhui, China
| | - Jia Wang
- Department of Pharmacology, School of Pharmaceutical Sciences,
Anhui Medical University, Hefei 230032, Anhui, China
| | - Xianbing Bai
- Department of Pharmacology, School of Pharmaceutical Sciences,
Anhui Medical University, Hefei 230032, Anhui, China
| | - Chenlei Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences,
Anhui Medical University, Hefei 230032, Anhui, China
| | - Xinyue Luan
- Department of Pharmacology, School of Pharmaceutical Sciences,
Anhui Medical University, Hefei 230032, Anhui, China
| | - Huailong Pei
- Department of Pharmacology, School of Pharmaceutical Sciences,
Anhui Medical University, Hefei 230032, Anhui, China
| | - Huan Liu
- Department of Pharmacology, School of Pharmaceutical Sciences,
Anhui Medical University, Hefei 230032, Anhui, China
| | - Xinyou Lv
- Department of Psychology, School of Humanities and Social Sciences,
University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Xiangming Ye
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital,
Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Binliang Tang
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital,
Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Ming Chen
- Department of Pharmacology, School of Pharmaceutical Sciences,
Anhui Medical University, Hefei 230032, Anhui, China
| |
Collapse
|
2
|
Meyer-Acosta KK, Diaz-Guerra E, Varma P, Aruk A, Mirsadeghi S, Muniz-Perez A, Rafati Y, Hosseini A, Nieto-Estevez V, Giugliano M, Navara C, Hsieh J. APOE4 impacts cortical neurodevelopment and alters network formation in human brain organoids. Stem Cell Reports 2025:102537. [PMID: 40541173 DOI: 10.1016/j.stemcr.2025.102537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 05/20/2025] [Accepted: 05/21/2025] [Indexed: 06/22/2025] Open
Abstract
Apolipoprotein E4 (APOE4) is the leading genetic risk factor for Alzheimer's disease. While most studies examine the role of APOE4 in aging, APOE4 causes persistent changes in brain structure as early as infancy and is associated with altered functional connectivity that extends beyond adolescence. Here, we used human induced pluripotent stem cell-derived cortical and ganglionic eminence organoids (COs and GEOs) to examine APOE4's influence during the development of cortical excitatory and inhibitory neurons. We show that APOE4 reduces cortical neurons and increases glia by promoting gliogenic transcriptional programs. In contrast, APOE4 increases proliferation and differentiation of GABAergic progenitors resulting in early and persistent increases in GABAergic neurons. Multi-electrode array recordings in assembloids revealed that APOE4 disrupts neural network function resulting in heightened excitability and synchronicity. Together, our data provide new insights on how APOE4 influences cortical neurodevelopmental processes and the establishment of functional networks.
Collapse
Affiliation(s)
- Karina K Meyer-Acosta
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Eva Diaz-Guerra
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Parul Varma
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Adyasha Aruk
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Sara Mirsadeghi
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Aranis Muniz-Perez
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Yousef Rafati
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Ali Hosseini
- International School of Advanced Studies, Neuroscience Area, V. Bonomea 265, 34136 Trieste, Italy
| | - Vanesa Nieto-Estevez
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Michele Giugliano
- Department of Biomedical, Metabolic & Neural Sciences, University of Modena and Reggio Emilia, V. Campi 287, 41125 Modena, Italy
| | - Christopher Navara
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA; Stem Cell Core, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Jenny Hsieh
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
3
|
Ikebara JM, Jorge RS, Marinho LSR, Higa GSV, Adhikari A, Reis FMCV, Borges FS, Ulrich H, Takada SH, De Pasquale R, Kihara AH. Hippocampal Interneurons Shape Spatial Coding Alterations in Neurological Disorders. Mol Neurobiol 2025:10.1007/s12035-025-05020-2. [PMID: 40392508 DOI: 10.1007/s12035-025-05020-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 04/29/2025] [Indexed: 05/22/2025]
Abstract
Hippocampal interneurons (INs) play a fundamental role in regulating neural oscillations, modulating excitatory circuits, and shaping spatial representation. While historically overshadowed by excitatory pyramidal cells in spatial coding research, recent advances have demonstrated that inhibitory INs not only coordinate network dynamics but also contribute directly to spatial information processing. This review aims to provide a novel integrative perspective on how distinct IN subtypes participate in spatial coding and how their dysfunction contributes to cognitive deficits in neurological disorders such as epilepsy, Alzheimer's disease (AD), traumatic brain injury (TBI), and cerebral hypoxia-ischemia. We synthesize recent findings demonstrating that different IN classes-including parvalbumin (PV)-, somatostatin (SST)-, cholecystokinin (CCK)-, and calretinin (CR)-expressing neurons-exhibit spatially selective activity, challenging traditional views of spatial representation, and influence memory consolidation through network-level interactions. By leveraging cutting-edge techniques such as in vivo calcium imaging and optogenetics, new evidence suggests that INs encode spatial information with a level of specificity previously attributed only to pyramidal cells. Furthermore, we investigate the impact of inhibitory circuit dysfunction in neurological disorders, examining how disruptions in interneuronal activity lead to impaired theta-gamma coupling, altered sharp wave ripples, and destabilized place cell representations, ultimately resulting in spatial memory deficits. This review advances the field by shifting the focus from pyramidal-centered models to a more nuanced understanding of the hippocampal network, emphasizing the active role of INs in spatial coding. By highlighting the translational potential of targeting inhibitory circuits for therapeutic interventions, we propose novel strategies for restoring hippocampal network function in neurological conditions. Readers will gain a comprehensive understanding of the emerging role of INs in spatial representation and the critical implications of their dysfunction, paving the way for future research on interneuron-targeted treatments for cognitive disorders.
Collapse
Affiliation(s)
- Juliane Midori Ikebara
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Renata Silva Jorge
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
| | - Luciana Simões Rafagnin Marinho
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
| | - Guilherme Shigueto Vilar Higa
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Avishek Adhikari
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Fernando M C V Reis
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Fernando S Borges
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, NY, 11203, USA
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Silvia Honda Takada
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
| | - Roberto De Pasquale
- Neurophysiology Laboratory, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Alexandre Hiroaki Kihara
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil.
| |
Collapse
|
4
|
Craig GE, Ramos L, Essig SR, Eagles NJ, Jaffe AE, Martinowich K, Hallock HL. Stimulation of Locus Ceruleus Inputs to the Prelimbic Cortex in Mice Induces Cell Type-Specific Expression of the Apoe Gene. eNeuro 2024; 11:ENEURO.0328-24.2024. [PMID: 39632090 PMCID: PMC11675532 DOI: 10.1523/eneuro.0328-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024] Open
Abstract
The medial frontal cortex (mFC) and locus ceruleus (LC) are two brain areas that have been implicated in a range of cognitive phenomena, such as attention, memory, and decision-making. Regulators of these brain regions at the molecular level are not well understood but might help to elucidate underlying mechanisms of disorders that present with deficits in these cognitive domains. To probe this, we used chemogenetic stimulation of neurons in the LC with axonal projections to the prelimbic subregion (PrL) of the mFC and subsequent bulk RNA sequencing from the mouse PrL. We found that stimulation of this circuit caused an increase in transcription of a host of genes, including the Apoe gene. To investigate cell type-specific expression of Apoe in the PrL, we used a dual-virus approach to express either the excitatory DREADD receptor hM3Dq in LC neurons with projections to the PrL or a control virus and found that increases in Apoe expression in the PrL following depolarization of LC inputs is enriched in GABAergic neurons in a sex-dependent manner. The results of these experiments yield insights into how Apoe expression affects function in a cortical microcircuit that is important for attention, memory, and decision-making and point to interneuron-specific expression of Apoe as a potential biomarker for circuit function in disorders such as attention-deficit hyperactivity disorder, schizophrenia, and Alzheimer's disease.
Collapse
Affiliation(s)
| | - Lizbeth Ramos
- Neuroscience Program, Lafayette College, Easton, Pennsylvania 18042
| | - Samuel R Essig
- Neuroscience Program, Lafayette College, Easton, Pennsylvania 18042
| | - Nicholas J Eagles
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland 21205
| | - Andrew E Jaffe
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland 21205
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland 21205
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
- Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
- The Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, Maryland 21205
| | - Henry L Hallock
- Neuroscience Program, Lafayette College, Easton, Pennsylvania 18042
| |
Collapse
|
5
|
Loix M, Vanherle S, Turri M, Kemp S, Fernandes KJL, Hendriks JJA, Bogie JFJ. Stearoyl-CoA desaturase-1: a potential therapeutic target for neurological disorders. Mol Neurodegener 2024; 19:85. [PMID: 39563397 PMCID: PMC11575020 DOI: 10.1186/s13024-024-00778-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024] Open
Abstract
Disturbances in the fatty acid lipidome are increasingly recognized as key drivers in the progression of various brain disorders. In this review article, we delve into the impact of Δ9 fatty acid desaturases, with a particular focus on stearoyl-CoA desaturase-1 (SCD1), within the setting of neuroinflammation, neurodegeneration, and brain repair. Over the past years, it was established that inhibition or deficiency of SCD1 not only suppresses neuroinflammation but also protects against neurodegeneration in conditions such as multiple sclerosis, Alzheimer's disease, and Parkinson's disease. This protective effect is achieved through different mechanisms including enhanced remyelination, reversal of synaptic and cognitive impairments, and mitigation of α-synuclein toxicity. Intriguingly, metabolic rerouting of fatty acids via SCD1 improves the pathology associated with X-linked adrenoleukodystrophy, suggesting context-dependent benign and harmful effects of SCD1 inhibition in the brain. Here, we summarize and discuss the cellular and molecular mechanisms underlying both the beneficial and detrimental effects of SCD1 in these neurological disorders. We explore commonalities and distinctions, shedding light on potential therapeutic challenges. Additionally, we touch upon future research directions that promise to deepen our understanding of SCD1 biology in brain disorders and potentially enhance the clinical utility of SCD1 inhibitors.
Collapse
Affiliation(s)
- Melanie Loix
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Sam Vanherle
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Marta Turri
- Research Center on Aging, CIUSSS de l'Estrie-CHUS, Sherbrooke, Canada
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam Neuroscience, Amsterdam UMC Location University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, NH, Netherlands
| | - Karl J L Fernandes
- Research Center on Aging, CIUSSS de l'Estrie-CHUS, Sherbrooke, Canada
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | - Jerome J A Hendriks
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Jeroen F J Bogie
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium.
- University MS Center Hasselt, Pelt, Belgium.
| |
Collapse
|
6
|
Raber J, Chaudhari M, De la Torre A, Holden S, Kessler K, Glaeser B, Lenarczyk M, Leonard SW, Borg A, Kwok A, Patel C, Kronenberg A, Olsen CM, Willey JS, Morré J, Choi J, Stevens JF, Bobe G, Minnier J, Baker J. Effects of 5-ion 6-beam sequential irradiation in the presence and absence of hindlimb or control hindlimb unloading on behavioral performances and plasma metabolic pathways of Fischer 344 rats. Front Physiol 2024; 15:1486767. [PMID: 39605860 PMCID: PMC11598337 DOI: 10.3389/fphys.2024.1486767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/07/2024] [Indexed: 11/29/2024] Open
Abstract
Introduction Effects and interactions between different spaceflight stressors are expected to be experienced by crew on missions when exposed to microgravity and galactic cosmic rays (GCRs). One of the limitations of previous studies on simulated weightlessness using hindlimb unloading (HU) is that a control HU condition was not included. Methods We characterized the behavioral performance of male Fischer rats 2 months after sham or total body irradiation with a simplified 5-ion 6-mixed-beam exposure representative of GCRs in the absence or presence of HU. Six months later, the plasma, hippocampus, and cortex were processed to determine whether the behavioral effects were associated with long-term alterations in the metabolic pathways. Results In the open field without and with objects, interactions were observed for radiation × HU. In the plasma of animals that were not under the HU or control HU condition, the riboflavin metabolic pathway was affected most for sham irradiation vs. 0.75 Gy exposure. Analysis of the effects of control HU on plasma in the sham-irradiated animals showed that the alanine, aspartate, glutamate, riboflavin, and glutamine metabolisms as well as arginine biosynthesis were affected. The effects of control HU on the hippocampus in the sham-irradiated animals showed that the phenylalanine, tyrosine, and tryptophan pathway was affected the most. Analysis of effects of 0.75 Gy irradiation on the cortex of control HU animals showed that the glutamine and glutamate metabolic pathway was affected similar to the hippocampus, while the riboflavin pathway was affected in animals that were not under the control HU condition. The effects of control HU on the cortex in sham-irradiated animals showed that the riboflavin metabolic pathway was affected. Animals receiving 0.75 Gy of irradiation showed impaired glutamine and glutamate metabolic pathway, whereas animals receiving 1.5 Gy of irradiation showed impaired riboflavin metabolic pathways. A total of 21 plasma metabolites were correlated with the behavioral measures, indicating that plasma and brain biomarkers associated with behavioral performance are dependent on the environmental conditions experienced. Discussion Phenylalanine, tyrosine, and tryptophan metabolism as well as phenylalanine and tryptophan as plasma metabolites are biomarkers that can be considered for spaceflight as they were revealed in both Fischer and WAG/Rij rats exposed to simGCRsim and/or HU.
Collapse
Affiliation(s)
- Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
- Departments of Neurology, Psychiatry, and Radiation Medicine, Division of Neuroscience ONPRC, Oregon Health and Science University, Portland, OR, United States
- College of Pharmacy, Oregon State University, Corvallis, OR, United States
| | - Mitali Chaudhari
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Alexis De la Torre
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Sarah Holden
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Kat Kessler
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Breanna Glaeser
- Neuroscience Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Marek Lenarczyk
- Radiation Biosciences laboratory, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Scott Willem Leonard
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Alexander Borg
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Andy Kwok
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Chirayu Patel
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Amy Kronenberg
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Christopher M. Olsen
- Neuroscience Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jeffrey S. Willey
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Jeffrey Morré
- Mass Spectrometry Center, Oregon State University, Corvallis, OR, United States
| | - Jaewoo Choi
- Mass Spectrometry Center, Oregon State University, Corvallis, OR, United States
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Jan Frederik Stevens
- College of Pharmacy, Oregon State University, Corvallis, OR, United States
- Mass Spectrometry Center, Oregon State University, Corvallis, OR, United States
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Gerd Bobe
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
- Department of Animal & Rangeland Sciences, Oregon State University, Corvallis, OR, United States
| | - Jessica Minnier
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - John Baker
- Radiation Biosciences laboratory, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
7
|
Sandoval KE, Witt KA. Somatostatin: Linking Cognition and Alzheimer Disease to Therapeutic Targeting. Pharmacol Rev 2024; 76:1291-1325. [PMID: 39013601 PMCID: PMC11549939 DOI: 10.1124/pharmrev.124.001117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024] Open
Abstract
Over 4 decades of research support the link between Alzheimer disease (AD) and somatostatin [somatotropin-releasing inhibitory factor (SRIF)]. SRIF and SRIF-expressing neurons play an essential role in brain function, modulating hippocampal activity and memory formation. Loss of SRIF and SRIF-expressing neurons in the brain rests at the center of a series of interdependent pathological events driven by amyloid-β peptide (Aβ), culminating in cognitive decline and dementia. The connection between the SRIF and AD further extends to the neuropsychiatric symptoms, seizure activity, and inflammation, whereas preclinical AD investigations show SRIF or SRIF receptor agonist administration capable of enhancing cognition. SRIF receptor subtype-4 activation in particular presents unique attributes, with the potential to mitigate learning and memory decline, reduce comorbid symptoms, and enhance enzymatic degradation of Aβ in the brain. Here, we review the links between SRIF and AD along with the therapeutic implications. SIGNIFICANCE STATEMENT: Somatostatin and somatostatin-expressing neurons in the brain are extensively involved in cognition. Loss of somatostatin and somatostatin-expressing neurons in Alzheimer disease rests at the center of a series of interdependent pathological events contributing to cognitive decline and dementia. Targeting somatostatin-mediated processes has significant therapeutic potential for the treatment of Alzheimer disease.
Collapse
Affiliation(s)
- Karin E Sandoval
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| | - Ken A Witt
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| |
Collapse
|
8
|
Meyer-Acosta KK, Diaz-Guerra E, Varma P, Aruk A, Mirsadeghi S, Perez AM, Rafati Y, Hosseini A, Nieto-Estevez V, Giugliano M, Navara C, Hsieh J. APOE4 impacts cortical neurodevelopment and alters network formation in human brain organoids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617044. [PMID: 39416105 PMCID: PMC11482793 DOI: 10.1101/2024.10.07.617044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Apolipoprotein E4 ( APOE4 ) is the leading genetic risk factor for Alzheimer's disease. While most studies examine the role of APOE4 in aging, imaging, and cognitive assessments reveal that APOE4 influences brain structure and function as early as infancy. Here, we examined human-relevant cellular phenotypes across neurodevelopment using induced pluripotent stem cell (iPSC) derived cortical and ganglionic eminence organoids (COs and GEOs). In COs, we showed that APOE4 decreased BRN2+ and SATB2+ cortical neurons, increased astrocytes and outer radial glia, and was associated with increased cell death and dysregulated GABA-related gene expression. In GEOs, APOE4 accelerated maturation of neural progenitors and neurons. Multi-electrode array recordings in assembloids revealed that APOE4 disrupted network formation and altered response to GABA, resulting in heightened excitability and synchronicity. Together, our data provides new insights into how APOE4 may influence cortical neurodevelopmental processes and network formation in the human brain.
Collapse
|
9
|
Geigenmüller JN, Tari AR, Wisloff U, Walker TL. The relationship between adult hippocampal neurogenesis and cognitive impairment in Alzheimer's disease. Alzheimers Dement 2024; 20:7369-7383. [PMID: 39166771 PMCID: PMC11485317 DOI: 10.1002/alz.14179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/14/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024]
Abstract
Neurogenesis persists throughout adulthood in the hippocampus and contributes to specific cognitive functions. In Alzheimer's disease (AD), the hippocampus is affected by pathology and functional impairment early in the disease. Human AD patients have reduced adult hippocampal neurogenesis (AHN) levels compared to age-matched healthy controls. Similarly, rodent AD models show a decrease in AHN before the onset of the classical hallmarks of AD pathology. Conversely, enhancement of AHN can protect against AD pathology and ameliorate memory deficits in both rodents and humans. Therefore, impaired AHN may be a contributing factor of AD-associated cognitive decline, rather than an effect of it. In this review we outline the regulation and function of AHN in healthy individuals, and highlight the relationship between AHN dysfunction and cognitive impairments in AD. The existence of AHN in humans and its relevance in AD patients will also be discussed, with an outlook toward future research directions. HIGHLIGHTS: Adult hippocampal neurogenesis occurs in the brains of mammals including humans. Adult hippocampal neurogenesis is reduced in Alzheimer's disease in humans and animal models.
Collapse
Affiliation(s)
| | - Atefe R. Tari
- The Cardiac Exercise Research Group at Department of Circulation and Medical ImagingFaculty of Medicine and Health SciencesNorwegian University of Science and Technology (NTNU)TrondheimNorway
- Department of Neurology and Clinical NeurophysiologySt. Olavs University Hospital, Trondheim University HospitalTrondheimNorway
| | - Ulrik Wisloff
- The Cardiac Exercise Research Group at Department of Circulation and Medical ImagingFaculty of Medicine and Health SciencesNorwegian University of Science and Technology (NTNU)TrondheimNorway
| | - Tara L. Walker
- Clem Jones Centre for Ageing Dementia ResearchQueensland Brain InstituteThe University of QueenslandBrisbaneAustralia
| |
Collapse
|
10
|
Christensen A, McGill CJ, Qian W, Pike CJ. Effects of obesogenic diet and 17β-estradiol in female mice with APOE 3/3, 3/4, and 4/4 genotypes. Front Aging Neurosci 2024; 16:1415072. [PMID: 39347015 PMCID: PMC11427389 DOI: 10.3389/fnagi.2024.1415072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/26/2024] [Indexed: 10/01/2024] Open
Abstract
The main genetic risk factor for Alzheimer's disease (AD) is the apolipoprotein E ε4 allele (APOE4). AD risk associated with APOE4 disproportionately affects women. Furthermore, human and rodent studies indicate that the cognitive deficits associated with APOE4 are greater in females. One modifiable AD risk factor is obesity during middle age. Given that approximately two-thirds of US adults are overweight, it is important to understand how obesity affects AD risk, how it interacts with APOE4, and the extent to which its detrimental effects can be mitigated with therapeutics. One intervention study for women is estrogen-based hormone therapy, which can exert numerous health benefits when administered in early middle age. No experimental studies have examined the interactions among APOE4, obesity, and hormone therapy in aging females. To begin to explore these issues, we considered how obesity outcomes are affected by treatment with estradiol at the onset of middle age in female mice with human APOE3 and APOE4. Furthermore, to explore how gene dosage affects outcomes, we compared mice homozygous for APOE3 (3/3) and homozygous (4/4) or hemizygous (3/4) for APOE4. Mice were examined over a 4-month period that spans the transition into reproductive senescence, a normal age-related change that models many aspects of human perimenopause. Beginning at 5 months of age, mice were maintained on a control diet (10% fat) or high-fat diet (HFD; 60% fat). After 8 weeks, by which time obesity was present in all HFD groups, mice were implanted with an estradiol or vehicle capsule that was maintained for the final 8 weeks. Animals were assessed on a range of metabolic and neural measures. Overall, APOE4 was associated with poorer metabolic function and cognitive performance. However, an obesogenic diet induced relatively greater impairments in metabolic function and cognitive performance in APOE3/3 mice. Estradiol treatment improved metabolic and cognitive outcomes across all HFD groups, with APOE4/4 generally exhibiting the greatest benefit. APOE3/4 mice were intermediate to the homozygous genotypes on many measures but also exhibited unique profiles. Together, these findings highlight the importance of the APOE genotype as a modulator of the risks associated with obesity and the beneficial outcomes of estradiol.
Collapse
Affiliation(s)
| | | | | | - Christian J. Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
11
|
Craig GE, Ramos L, Essig SR, Eagles NJ, Jaffe AE, Martinowich K, Hallock HL. Stimulation of locus coeruleus inputs to the frontal cortex in mice induces cell type-specific expression of the Apoe gene. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.22.604695. [PMID: 39091890 PMCID: PMC11291023 DOI: 10.1101/2024.07.22.604695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Deficits in attention are common across a range of neuropsychiatric disorders. A multitude of brain regions, including the frontal cortex (FC) and locus coeruleus (LC), have been implicated in attention. Regulators of these brain regions at the molecular level are not well understood, but might elucidate underlying mechanisms of disorders with attentional deficits. To probe this, we used chemogenetic stimulation of neurons in the LC with axonal projections to the FC, and subsequent bulk RNA-sequencing from the mouse FC. We found that stimulation of this circuit caused an increase in transcription of the Apoe gene. To investigate cell type-specific expression of Apoe in the FC, we used a dual-virus approach to express either the excitatory DREADD receptor hM3Dq in LC neurons with projections to the FC, or a control virus, and found that increases in Apoe expression in the FC following depolarization of LC inputs is enriched in GABAergic neurons in a sex-dependent manner. The results of these experiments yield insights into how Apoe expression affects function in cortical microcircuits that are important for attention-guided behavior, and point to interneuron-specific expression of Apoe as a potential target for the amelioration of attention symptoms in disorders such as attention-deficit hyperactivity disorder (ADHD), schizophrenia, and Alzheimer's disease (AD).
Collapse
Affiliation(s)
| | - Lizbeth Ramos
- Neuroscience Program, Lafayette College, Easton, PA, 18042, USA
| | - Samuel R. Essig
- Neuroscience Program, Lafayette College, Easton, PA, 18042, USA
| | - Nicholas J. Eagles
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Andrew E. Jaffe
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- The Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, 21205, USA
| | | |
Collapse
|
12
|
Charou D, Rogdakis T, Latorrata A, Valcarcel M, Papadogiannis V, Athanasiou C, Tsengenes A, Papadopoulou MA, Lypitkas D, Lavigne MD, Katsila T, Wade RC, Cader MZ, Calogeropoulou T, Gravanis A, Charalampopoulos I. Comprehensive characterization of the neurogenic and neuroprotective action of a novel TrkB agonist using mouse and human stem cell models of Alzheimer's disease. Stem Cell Res Ther 2024; 15:200. [PMID: 38971770 PMCID: PMC11227723 DOI: 10.1186/s13287-024-03818-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 06/26/2024] [Indexed: 07/08/2024] Open
Abstract
BACKGROUND Neural stem cell (NSC) proliferation and differentiation in the mammalian brain decreases to minimal levels postnatally. Nevertheless, neurogenic niches persist in the adult cortex and hippocampus in rodents, primates and humans, with adult NSC differentiation sharing key regulatory mechanisms with development. Adult neurogenesis impairments have been linked to Alzheimer's disease (AD) pathology. Addressing these impairments by using neurotrophic factors is a promising new avenue for therapeutic intervention based on neurogenesis. However, this possibility has been hindered by technical difficulties of using in-vivo models to conduct screens, including working with scarce NSCs in the adult brain and differences between human and mouse models or ethical limitations. METHODS Here, we use a combination of mouse and human stem cell models for comprehensive in-vitro characterization of a novel neurogenic compound, focusing on the brain-derived neurotrophic factor (BDNF) pathway. The ability of ENT-A011, a steroidal dehydroepiandrosterone derivative, to activate the tyrosine receptor kinase B (TrkB) receptor was tested through western blotting in NIH-3T3 cells and its neurogenic and neuroprotective action were assessed through proliferation, cell death and Amyloid-β (Aβ) toxicity assays in mouse primary adult hippocampal NSCs, mouse embryonic cortical NSCs and neural progenitor cells (NPCs) differentiated from three human induced pluripotent stem cell lines from healthy and AD donors. RNA-seq profiling was used to assess if the compound acts through the same gene network as BDNF in human NPCs. RESULTS ENT-A011 was able to increase proliferation of mouse primary adult hippocampal NSCs and embryonic cortical NSCs, in the absence of EGF/FGF, while reducing Aβ-induced cell death, acting selectively through TrkB activation. The compound was able to increase astrocytic gene markers involved in NSC maintenance, protect hippocampal neurons from Αβ toxicity and prevent synapse loss after Aβ treatment. ENT-A011 successfully induces proliferation and prevents cell death after Aβ toxicity in human NPCs, acting through a core gene network shared with BDNF as shown through RNA-seq. CONCLUSIONS Our work characterizes a novel BDNF mimetic with preferable pharmacological properties and neurogenic and neuroprotective actions in Alzheimer's disease via stem cell-based screening, demonstrating the promise of stem cell systems for short-listing competitive candidates for further testing.
Collapse
Affiliation(s)
- Despoina Charou
- Department of Pharmacology, Medical School, University of Crete, 71003, Heraklion, Greece
- Foundation for Research and Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology and Biotechnology, 70013, Heraklion, Greece
| | - Thanasis Rogdakis
- Department of Pharmacology, Medical School, University of Crete, 71003, Heraklion, Greece
- Foundation for Research and Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology and Biotechnology, 70013, Heraklion, Greece
| | - Alessia Latorrata
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635, Athens, Greece
| | - Maria Valcarcel
- Innovative Technologies in Biological Systems SL (INNOPROT), 48160, Derio, Bizkaia, Spain
| | - Vasileios Papadogiannis
- Hellenic Centre for Marine Research (HCMR), Institute of Marine Biology Biotechnology and Aquaculture (IMBBC), Heraklion, Crete, Greece
| | - Christina Athanasiou
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), 69118, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
- Heidelberg Biosciences International Graduate School, Heidelberg University, 69120, Heidelberg, Germany
| | - Alexandros Tsengenes
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), 69118, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
- Heidelberg Biosciences International Graduate School, Heidelberg University, 69120, Heidelberg, Germany
| | - Maria Anna Papadopoulou
- Department of Pharmacology, Medical School, University of Crete, 71003, Heraklion, Greece
- Foundation for Research and Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology and Biotechnology, 70013, Heraklion, Greece
| | - Dimitrios Lypitkas
- Department of Pharmacology, Medical School, University of Crete, 71003, Heraklion, Greece
- Foundation for Research and Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology and Biotechnology, 70013, Heraklion, Greece
| | - Matthieu D Lavigne
- Foundation for Research and Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology and Biotechnology, 70013, Heraklion, Greece
| | - Theodora Katsila
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635, Athens, Greece
| | - Rebecca C Wade
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), 69118, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, 69120, Heidelberg, Germany
- Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, 69120, Heidelberg, Germany
| | - M Zameel Cader
- Translational Molecular Neuroscience Group, Dorothy Crowfoot Hodgkin Building, Kavli Institute for Nanoscience, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | - Achille Gravanis
- Department of Pharmacology, Medical School, University of Crete, 71003, Heraklion, Greece
- Foundation for Research and Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology and Biotechnology, 70013, Heraklion, Greece
| | - Ioannis Charalampopoulos
- Department of Pharmacology, Medical School, University of Crete, 71003, Heraklion, Greece.
- Foundation for Research and Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology and Biotechnology, 70013, Heraklion, Greece.
| |
Collapse
|
13
|
Kunoh S, Nakashima H, Nakashima K. Epigenetic Regulation of Neural Stem Cells in Developmental and Adult Stages. EPIGENOMES 2024; 8:22. [PMID: 38920623 PMCID: PMC11203245 DOI: 10.3390/epigenomes8020022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/18/2024] [Accepted: 05/31/2024] [Indexed: 06/27/2024] Open
Abstract
The development of the nervous system is regulated by numerous intracellular molecules and cellular signals that interact temporally and spatially with the extracellular microenvironment. The three major cell types in the brain, i.e., neurons and two types of glial cells (astrocytes and oligodendrocytes), are generated from common multipotent neural stem cells (NSCs) throughout life. However, NSCs do not have this multipotentiality from the beginning. During cortical development, NSCs sequentially obtain abilities to differentiate into neurons and glial cells in response to combinations of spatiotemporally modulated cell-intrinsic epigenetic alterations and extrinsic factors. After the completion of brain development, a limited population of NSCs remains in the adult brain and continues to produce neurons (adult neurogenesis), thus contributing to learning and memory. Many biological aspects of brain development and adult neurogenesis are regulated by epigenetic changes via behavioral control of NSCs. Epigenetic dysregulation has also been implicated in the pathogenesis of various brain diseases. Here, we present recent advances in the epigenetic regulation of NSC behavior and its dysregulation in brain disorders.
Collapse
Affiliation(s)
| | - Hideyuki Nakashima
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan;
| | - Kinichi Nakashima
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan;
| |
Collapse
|
14
|
Casciati A, Pasquali E, De Stefano I, Braga-Tanaka I, Tanaka S, Mancuso M, Antonelli F, Pazzaglia S. Role of Apolipoprotein E in the Hippocampus and Its Impact following Ionizing Radiation Exposure. Cells 2024; 13:899. [PMID: 38891031 PMCID: PMC11171511 DOI: 10.3390/cells13110899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
Apolipoprotein E (ApoE) is a lipid carrier in both the peripheral and the central nervous systems (CNSs). Lipid-loaded ApoE lipoprotein particles bind to several cell surface receptors to support membrane homeostasis and brain injury repair. In the brain, ApoE is produced predominantly by astrocytes, but it is also abundantly expressed in most neurons of the CNS. In this study, we addressed the role of ApoE in the hippocampus in mice, focusing on its role in response to radiation injury. To this aim, 8-week-old, wild-type, and ApoE-deficient (ApoE-/-) female mice were acutely whole-body irradiated with 3 Gy of X-rays (0.89 Gy/min), then sacrificed 150 days post-irradiation. In addition, age-matching ApoE-/- females were chronically whole-body irradiated (20 mGy/d, cumulative dose of 3 Gy) for 150 days at the low dose-rate facility at the Institute of Environmental Sciences (IES), Rokkasho, Japan. To seek for ApoE-dependent modification during lineage progression from neural stem cells to neurons, we have evaluated the cellular composition of the dentate gyrus in unexposed and irradiated mice using stage-specific markers of adult neurogenesis. Our findings indicate that ApoE genetic inactivation markedly perturbs adult hippocampal neurogenesis in unexposed and irradiated mice. The effect of ApoE inactivation on the expression of a panel of miRNAs with an established role in hippocampal neurogenesis, as well as its transcriptional consequences in their target genes regulating neurogenic program, have also been analyzed. Our data show that the absence of ApoE-/- also influences synaptic functionality and integration by interfering with the regulation of mir-34a, mir-29b, and mir-128b, leading to the downregulation of synaptic markers PSD95 and synaptophysin mRNA. Finally, compared to acute irradiation, chronic exposure of ApoE null mice yields fewer consequences except for the increased microglia-mediated neuroinflammation. Exploring the function of ApoE in the hippocampus could have implications for developing therapeutic approaches to alleviate radiation-induced brain injury.
Collapse
Affiliation(s)
- Arianna Casciati
- Division Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (A.C.); (E.P.); (I.D.S.); (M.M.)
| | - Emanuela Pasquali
- Division Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (A.C.); (E.P.); (I.D.S.); (M.M.)
| | - Ilaria De Stefano
- Division Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (A.C.); (E.P.); (I.D.S.); (M.M.)
| | - Ignacia Braga-Tanaka
- Department of Radiobiology, Institute for Environmental Sciences, Rokkasho 039-3212, Japan; (I.B.-T.); (S.T.)
| | - Satoshi Tanaka
- Department of Radiobiology, Institute for Environmental Sciences, Rokkasho 039-3212, Japan; (I.B.-T.); (S.T.)
| | - Mariateresa Mancuso
- Division Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (A.C.); (E.P.); (I.D.S.); (M.M.)
| | - Francesca Antonelli
- Division Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (A.C.); (E.P.); (I.D.S.); (M.M.)
| | - Simonetta Pazzaglia
- Division Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (A.C.); (E.P.); (I.D.S.); (M.M.)
| |
Collapse
|
15
|
Jeong SP, Sharma N, An SSA. Role of Calcitriol and Vitamin D Receptor ( VDR) Gene Polymorphisms in Alzheimer's Disease. Int J Mol Sci 2024; 25:4806. [PMID: 38732025 PMCID: PMC11084202 DOI: 10.3390/ijms25094806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 02/21/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by amyloid beta (Aβ) buildup and neuronal degeneration. An association between low serum vitamin D levels and an increased risk of AD has been reported in several epidemiological studies. Calcitriol (1,25-dihydroxycholecalciferol) is the active form of vitamin D, and is generated in the kidney and many other tissues/organs, including the brain. It is a steroid hormone that regulates important functions like calcium/phosphorous levels, bone mineralization, and immunomodulation, indicating its broader systemic significance. In addition, calcitriol confers neuroprotection by mitigating oxidative stress and neuroinflammation, promoting the clearance of Aβ, myelin formation, neurogenesis, neurotransmission, and autophagy. The receptors to which calcitriol binds (vitamin D receptors; VDRs) to exert its effects are distributed over many organs and tissues, representing other significant roles of calcitriol beyond sustaining bone health. The biological effects of calcitriol are manifested through genomic (classical) and non-genomic actions through different pathways. The first is a slow genomic effect involving nuclear VDR directly affecting gene transcription. The association of AD with VDR gene polymorphisms relies on the changes in vitamin D consumption, which lowers VDR expression, protein stability, and binding affinity. It leads to the altered expression of genes involved in the neuroprotective effects of calcitriol. This review summarizes the neuroprotective mechanism of calcitriol and the role of VDR polymorphisms in AD, and might help develop potential therapeutic strategies and markers for AD in the future.
Collapse
Affiliation(s)
| | - Niti Sharma
- Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujung-gu, Seongnam-si 461-701, Republic of Korea
| | - Seong Soo A. An
- Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujung-gu, Seongnam-si 461-701, Republic of Korea
| |
Collapse
|
16
|
Patil SP, Kuehn BR. Discovery of Small Molecule Glycolytic Stimulants for Enhanced ApoE Lipidation in Alzheimer's Disease Cell Model. Pharmaceuticals (Basel) 2024; 17:491. [PMID: 38675451 PMCID: PMC11054693 DOI: 10.3390/ph17040491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/28/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by pathophysiological deposits of extracellular amyloid beta (Aβ) peptides and intracellular neurofibrillary tangles of tau. The central role of Aβ in AD pathology is well-established, with its increased deposition attributed mainly to its decreased cerebral clearance. Here, it is noteworthy that apolipoprotein E (ApoE), the most significant risk factor for AD, has been shown to play an isoform-specific role in clearing Aβ deposits (ApoE2 > ApoE3 > ApoE4), owing mainly to its lipidation status. In addition to the pathophysiological Aβ deposits, AD is also characterized by abnormal glucose metabolism, which is a distinct event preceding Aβ deposition. The present study established, for the first time, a possible link between these two major AD etiologies, with glucose metabolism directly influencing ApoE lipidation and its secretion by astrocytes expressing human ApoE4. Specifically, glucose dose-dependently activated liver X receptor (LXR), leading to elevated ABCA1 and ABCG1 protein levels and enhanced ApoE lipidation. Moreover, co-treatment with a glycolytic inhibitor significantly inhibited this LXR activation and subsequent ApoE lipidation, further supporting a central role of glucose metabolism in LXR activation leading to enhanced ApoE lipidation, which may help against AD through potential Aβ clearance. Therefore, we hypothesized that pharmacological agents that can target cellular energy metabolism, specifically aerobic glycolysis, may hold significant therapeutic potential against AD. In this context, the present study also led to the discovery of novel, small-molecule stimulants of astrocytic glucose metabolism, leading to significantly enhanced lipidation status of ApoE4 in astrocytic cells. Three such newly discovered compounds (lonidamine, phenformin, and berberine), owing to their promising cellular effect on the glycolysis-ApoE nexus, warrant further investigation in suitable in vivo models of AD.
Collapse
Affiliation(s)
- Sachin P. Patil
- NanoBio Lab, Widener University, Chester, PA 19013, USA
- Department of Chemical Engineering, Widener University, Chester, PA 19013, USA;
| | - Bella R. Kuehn
- Department of Chemical Engineering, Widener University, Chester, PA 19013, USA;
| |
Collapse
|
17
|
Almeida VN. Somatostatin and the pathophysiology of Alzheimer's disease. Ageing Res Rev 2024; 96:102270. [PMID: 38484981 DOI: 10.1016/j.arr.2024.102270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 03/09/2024] [Accepted: 03/09/2024] [Indexed: 03/28/2024]
Abstract
Among the central features of Alzheimer's disease (AD) progression are altered levels of the neuropeptide somatostatin (SST), and the colocalisation of SST-positive interneurons (SST-INs) with amyloid-β plaques, leading to cell death. In this theoretical review, I propose a molecular model for the pathogenesis of AD based on SST-IN hypofunction and hyperactivity. Namely, hypofunctional and hyperactive SST-INs struggle to control hyperactivity in medial regions in early stages, leading to axonal Aβ production through excessive presynaptic GABAB inhibition, GABAB1a/APP complex downregulation and internalisation. Concomitantly, excessive SST-14 release accumulates near SST-INs in the form of amyloids, which bind to Aβ to form toxic mixed oligomers. This leads to differential SST-IN death through excitotoxicity, further disinhibition, SST deficits, and increased Aβ release, fibrillation and plaque formation. Aβ plaques, hyperactive networks and SST-IN distributions thereby tightly overlap in the brain. Conversely, chronic stimulation of postsynaptic SST2/4 on gulutamatergic neurons by hyperactive SST-INs promotes intense Mitogen-Activated Protein Kinase (MAPK) p38 activity, leading to somatodendritic p-tau staining and apoptosis/neurodegeneration - in agreement with a near complete overlap between p38 and neurofibrillary tangles. This model is suitable to explain some of the principal risk factors and markers of AD progression, including mitochondrial dysfunction, APOE4 genotype, sex-dependent vulnerability, overactive glial cells, dystrophic neurites, synaptic/spine losses, inter alia. Finally, the model can also shed light on qualitative aspects of AD neuropsychology, especially within the domains of spatial and declarative (episodic, semantic) memory, under an overlying pattern of contextual indiscrimination, ensemble instability, interference and generalisation.
Collapse
Affiliation(s)
- Victor N Almeida
- Institute of Psychiatry, Faculty of Medicine, University of São Paulo (USP), Brazil; Faculty of Languages, Federal University of Minas Gerais (UFMG), Brazil.
| |
Collapse
|
18
|
Tan J, Tan YY, Ngian ZK, Chong SY, Rao VK, Wang JW, Zeng X, Ong CT. ApoE maintains neuronal integrity via microRNA and H3K27me3-mediated repression. iScience 2024; 27:109231. [PMID: 38439966 PMCID: PMC10909902 DOI: 10.1016/j.isci.2024.109231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 12/15/2023] [Accepted: 02/09/2024] [Indexed: 03/06/2024] Open
Abstract
ApoE regulates neurogenesis, although how it influences genetic programs remains elusive. Cortical neurons induced from isogenic control and ApoE-/- human neural stem cells (NSCs) recapitulated key transcriptomic signatures of in vivo counterparts identified from single-cell human midbrain. Surprisingly, ApoE expression in NSC and neural progenitor cells (NPCs) is not required for differentiation. Instead, ApoE prevents the over-proliferation of non-neuronal cells during extended neuronal culture when it is not expressed. Elevated miR-199a-5p level in ApoE-/- cells lowers the EZH1 protein and the repressive H3K27me3 mark, a phenotype rescued by miR-199a-5p steric inhibitor. Reduced H3K27me3 at genes linked to extracellular matrix organization and angiogenesis in ApoE-/- NPC correlates with their aberrant expression and phenotypes in neurons. Interestingly, the ApoE coding sequence, which contains many predicted miR-199a-5p binding sites, can repress miR-199a-5p without translating into protein. This suggests that ApoE maintains neurons integrity through the target-directed miRNA degradation of miR-199a-5p, imparting the H3K27me3-mediated repression of non-neuronal genes during differentiation.
Collapse
Affiliation(s)
- Jiazi Tan
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604, Singapore
| | - Yow-Yong Tan
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Zhen-Kai Ngian
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604, Singapore
| | - Suet-Yen Chong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Vinay Kumar Rao
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604, Singapore
- Department of Medical Genetics, JSS Medical College, JSS Academy of Higher Education and Research, Mysore 570015, India
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117609, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Xianmin Zeng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- RxCell Inc, Novato, CA 94945, USA
| | - Chin-Tong Ong
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| |
Collapse
|
19
|
Santiago RMM, Lopes-Dos-Santos V, Aery Jones EA, Huang Y, Dupret D, Tort ABL. Waveform-based classification of dentate spikes. Sci Rep 2024; 14:2989. [PMID: 38316828 PMCID: PMC10844627 DOI: 10.1038/s41598-024-53075-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 01/27/2024] [Indexed: 02/07/2024] Open
Abstract
Synchronous excitatory discharges from the entorhinal cortex (EC) to the dentate gyrus (DG) generate fast and prominent patterns in the hilar local field potential (LFP), called dentate spikes (DSs). As sharp-wave ripples in CA1, DSs are more likely to occur in quiet behavioral states, when memory consolidation is thought to take place. However, their functions in mnemonic processes are yet to be elucidated. The classification of DSs into types 1 or 2 is determined by their origin in the lateral or medial EC, as revealed by current source density (CSD) analysis, which requires recordings from linear probes with multiple electrodes spanning the DG layers. To allow the investigation of the functional role of each DS type in recordings obtained from single electrodes and tetrodes, which are abundant in the field, we developed an unsupervised method using Gaussian mixture models to classify such events based on their waveforms. Our classification approach achieved high accuracies (> 80%) when validated in 8 mice with DG laminar profiles. The average CSDs, waveforms, rates, and widths of the DS types obtained through our method closely resembled those derived from the CSD-based classification. As an example of application, we used the technique to analyze single-electrode LFPs from apolipoprotein (apo) E3 and apoE4 knock-in mice. We observed that the latter group, which is a model for Alzheimer's disease, exhibited wider DSs of both types from a young age, with a larger effect size for DS type 2, likely reflecting early pathophysiological alterations in the EC-DG network, such as hyperactivity. In addition to the applicability of the method in expanding the study of DS types, our results show that their waveforms carry information about their origins, suggesting different underlying network dynamics and roles in memory processing.
Collapse
Affiliation(s)
- Rodrigo M M Santiago
- Computational Neurophysiology Lab, Brain Institute, Federal University of Rio Grande do Norte, Natal, RN, 59078-900, Brazil.
| | - Vítor Lopes-Dos-Santos
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Emily A Aery Jones
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, 94158, USA
| | - David Dupret
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Adriano B L Tort
- Computational Neurophysiology Lab, Brain Institute, Federal University of Rio Grande do Norte, Natal, RN, 59078-900, Brazil
| |
Collapse
|
20
|
Blumenfeld J, Yip O, Kim MJ, Huang Y. Cell type-specific roles of APOE4 in Alzheimer disease. Nat Rev Neurosci 2024; 25:91-110. [PMID: 38191720 PMCID: PMC11073858 DOI: 10.1038/s41583-023-00776-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 01/10/2024]
Abstract
The ɛ4 allele of the apolipoprotein E gene (APOE), which translates to the APOE4 isoform, is the strongest genetic risk factor for late-onset Alzheimer disease (AD). Within the CNS, APOE is produced by a variety of cell types under different conditions, posing a challenge for studying its roles in AD pathogenesis. However, through powerful advances in research tools and the use of novel cell culture and animal models, researchers have recently begun to study the roles of APOE4 in AD in a cell type-specific manner and at a deeper and more mechanistic level than ever before. In particular, cutting-edge omics studies have enabled APOE4 to be studied at the single-cell level and have allowed the identification of critical APOE4 effects in AD-vulnerable cellular subtypes. Through these studies, it has become evident that APOE4 produced in various types of CNS cell - including astrocytes, neurons, microglia, oligodendrocytes and vascular cells - has diverse roles in AD pathogenesis. Here, we review these scientific advances and propose a cell type-specific APOE4 cascade model of AD. In this model, neuronal APOE4 emerges as a crucial pathological initiator and driver of AD pathogenesis, instigating glial responses and, ultimately, neurodegeneration. In addition, we provide perspectives on future directions for APOE4 research and related therapeutic developments in the context of AD.
Collapse
Affiliation(s)
- Jessica Blumenfeld
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Oscar Yip
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Min Joo Kim
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA.
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA.
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
21
|
Ji X, Peng X, Tang H, Pan H, Wang W, Wu J, Chen J, Wei N. Alzheimer's disease phenotype based upon the carrier status of the apolipoprotein E ɛ4 allele. Brain Pathol 2024; 34:e13208. [PMID: 37646624 PMCID: PMC10711266 DOI: 10.1111/bpa.13208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 08/05/2023] [Indexed: 09/01/2023] Open
Abstract
The apolipoprotein E ɛ4 allele (APOE4) is universally acknowledged as the most potent genetic risk factor for Alzheimer's disease (AD). APOE4 promotes the initiation and progression of AD. Although the underlying mechanisms are unclearly understood, differences in lipid-bound affinity among the three APOE isoforms may constitute the basis. The protein APOE4 isoform has a high affinity with triglycerides and cholesterol. A distinction in lipid metabolism extensively impacts neurons, microglia, and astrocytes. APOE4 carriers exhibit phenotypic differences from non-carriers in clinical examinations and respond differently to multiple treatments. Therefore, we hypothesized that phenotypic classification of AD patients according to the status of APOE4 carrier will help specify research and promote its use in diagnosing and treating AD. Recent reviews have mainly evaluated the differences between APOE4 allele carriers and non-carriers from gene to protein structures, clinical features, neuroimaging, pathology, the neural network, and the response to various treatments, and have provided the feasibility of phenotypic group classification based on APOE4 carrier status. This review will facilitate the application of APOE phenomics concept in clinical practice and promote further medical research on AD.
Collapse
Affiliation(s)
- Xiao‐Yu Ji
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
- Brain Function and Disease LaboratoryShantou University Medical CollegeGuangdongChina
| | - Xin‐Yuan Peng
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
| | - Hai‐Liang Tang
- Fudan University Huashan Hospital, Department of Neurosurgery, State Key Laboratory for Medical NeurobiologyInstitutes of Brain Science, Shanghai Medical College‐Fudan UniversityShanghaiChina
| | - Hui Pan
- Shantou Longhu People's HospitalShantouGuangdongChina
| | - Wei‐Tang Wang
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
| | - Jie Wu
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
- Brain Function and Disease LaboratoryShantou University Medical CollegeGuangdongChina
| | - Jian Chen
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
| | - Nai‐Li Wei
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
| |
Collapse
|
22
|
Hijazi S, Smit AB, van Kesteren RE. Fast-spiking parvalbumin-positive interneurons in brain physiology and Alzheimer's disease. Mol Psychiatry 2023; 28:4954-4967. [PMID: 37419975 PMCID: PMC11041664 DOI: 10.1038/s41380-023-02168-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/09/2023]
Abstract
Fast-spiking parvalbumin (PV) interneurons are inhibitory interneurons with unique morphological and functional properties that allow them to precisely control local circuitry, brain networks and memory processing. Since the discovery in 1987 that PV is expressed in a subset of fast-spiking GABAergic inhibitory neurons, our knowledge of the complex molecular and physiological properties of these cells has been expanding. In this review, we highlight the specific properties of PV neurons that allow them to fire at high frequency and with high reliability, enabling them to control network oscillations and shape the encoding, consolidation and retrieval of memories. We next discuss multiple studies reporting PV neuron impairment as a critical step in neuronal network dysfunction and cognitive decline in mouse models of Alzheimer's disease (AD). Finally, we propose potential mechanisms underlying PV neuron dysfunction in AD and we argue that early changes in PV neuron activity could be a causal step in AD-associated network and memory impairment and a significant contributor to disease pathogenesis.
Collapse
Affiliation(s)
- Sara Hijazi
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Ronald E van Kesteren
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
23
|
Nelson MR, Liu P, Agrawal A, Yip O, Blumenfeld J, Traglia M, Kim MJ, Koutsodendris N, Rao A, Grone B, Hao Y, Yoon SY, Xu Q, De Leon S, Choenyi T, Thomas R, Lopera F, Quiroz YT, Arboleda-Velasquez JF, Reiman EM, Mahley RW, Huang Y. The APOE-R136S mutation protects against APOE4-driven Tau pathology, neurodegeneration and neuroinflammation. Nat Neurosci 2023; 26:2104-2121. [PMID: 37957317 PMCID: PMC10689245 DOI: 10.1038/s41593-023-01480-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 10/04/2023] [Indexed: 11/15/2023]
Abstract
Apolipoprotein E4 (APOE4) is the strongest genetic risk factor for late-onset Alzheimer's disease (LOAD), leading to earlier age of clinical onset and exacerbating pathologies. There is a critical need to identify protective targets. Recently, a rare APOE variant, APOE3-R136S (Christchurch), was found to protect against early-onset AD in a PSEN1-E280A carrier. In this study, we sought to determine if the R136S mutation also protects against APOE4-driven effects in LOAD. We generated tauopathy mouse and human iPSC-derived neuron models carrying human APOE4 with the homozygous or heterozygous R136S mutation. We found that the homozygous R136S mutation rescued APOE4-driven Tau pathology, neurodegeneration and neuroinflammation. The heterozygous R136S mutation partially protected against APOE4-driven neurodegeneration and neuroinflammation but not Tau pathology. Single-nucleus RNA sequencing revealed that the APOE4-R136S mutation increased disease-protective and diminished disease-associated cell populations in a gene dose-dependent manner. Thus, the APOE-R136S mutation protects against APOE4-driven AD pathologies, providing a target for therapeutic development against AD.
Collapse
Affiliation(s)
- Maxine R Nelson
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Peng Liu
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Ayushi Agrawal
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA
| | - Oscar Yip
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Jessica Blumenfeld
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Michela Traglia
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA
| | - Min Joo Kim
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Nicole Koutsodendris
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Antara Rao
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Brian Grone
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Yanxia Hao
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Seo Yeon Yoon
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Qin Xu
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Samuel De Leon
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Tenzing Choenyi
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Reuben Thomas
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia de la Universidad de Antioquia, Medellin, Colombia
| | - Yakeel T Quiroz
- Grupo de Neurociencias de Antioquia de la Universidad de Antioquia, Medellin, Colombia
- Departments of Neurology and Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Joseph F Arboleda-Velasquez
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Eric M Reiman
- Banner Alzheimer's Institute, Phoenix, AZ, USA
- University of Arizona, Tucson, AZ, USA
| | - Robert W Mahley
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA.
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA.
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA.
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
24
|
Santiago RM, Lopes-dos-Santos V, Jones EAA, Huang Y, Dupret D, Tort AB. Waveform-based classification of dentate spikes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563826. [PMID: 37961150 PMCID: PMC10634814 DOI: 10.1101/2023.10.24.563826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Synchronous excitatory discharges from the entorhinal cortex (EC) to the dentate gyrus (DG) generate fast and prominent patterns in the hilar local field potential (LFP), called dentate spikes (DSs). As sharp-wave ripples in CA1, DSs are more likely to occur in quiet behavioral states, when memory consolidation is thought to take place. However, their functions in mnemonic processes are yet to be elucidated. The classification of DSs into types 1 or 2 is determined by their origin in the lateral or medial EC, as revealed by current source density (CSD) analysis, which requires recordings from linear probes with multiple electrodes spanning the DG layers. To allow the investigation of the functional role of each DS type in recordings obtained from single electrodes and tetrodes, which are abundant in the field, we developed an unsupervised method using Gaussian mixture models to classify such events based on their waveforms. Our classification approach achieved high accuracies (> 80%) when validated in 8 mice with DG laminar profiles. The average CSDs, waveforms, rates, and widths of the DS types obtained through our method closely resembled those derived from the CSD-based classification. As an example of application, we used the technique to analyze single-electrode LFPs from apolipoprotein (apo) E3 and apoE4 knock-in mice. We observed that the latter group, which is a model for Alzheimer's disease, exhibited wider DSs of both types from a young age, with a larger effect size for DS type 2, likely reflecting early pathophysiological alterations in the EC-DG network, such as hyperactivity. In addition to the applicability of the method in expanding the study of DS types, our results show that their waveforms carry information about their origins, suggesting different underlying network dynamics and roles in memory processing.
Collapse
Affiliation(s)
- Rodrigo M.M. Santiago
- Computational Neurophysiology Lab, Brain Institute, Federal University of Rio Grande do Norte, Natal, RN, 59078-900, Brazil
| | - Vítor Lopes-dos-Santos
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Emily A. Aery Jones
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - David Dupret
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Adriano B.L. Tort
- Computational Neurophysiology Lab, Brain Institute, Federal University of Rio Grande do Norte, Natal, RN, 59078-900, Brazil
| |
Collapse
|
25
|
Yuasa K, Hirosawa T, Soma D, Furutani N, Kameya M, Sano M, Kitamura K, Ueda M, Kikuchi M. Eyes-state-dependent alterations of magnetoencephalographic connectivity associated with delayed recall in Alzheimer's disease via graph theory approach. Front Psychiatry 2023; 14:1272120. [PMID: 37941968 PMCID: PMC10628524 DOI: 10.3389/fpsyt.2023.1272120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/10/2023] [Indexed: 11/10/2023] Open
Abstract
IntroductionAlzheimer’s disease (AD) is a neurodegenerative disorder characterized by memory impairment and cognitive decline. Electroencephalography (EEG) and magnetoencephalography (MEG) studies using graph theory show altered “Small-Worldness (SW)” properties in AD. This study aimed to investigate whether eye-state-dependent alterations in SW differ between patients with AD and healthy controls, considering the symptoms of AD.MethodsNineteen patients with AD and 24 healthy controls underwent MEG under different conditions (eyes-open [EO] and eyes-closed [EC]) and the Wechsler Memory Scale-Revised (WMS-R) with delayed recall. After the signal sources were mapped onto the Desikan–Killiany brain atlas, the statistical connectivity of five frequency bands (delta, theta, alpha, beta, and gamma) was calculated using the phase lag index (PLI), and binary graphs for each frequency band were constructed based on the PLI. Next, we measured SW as a graph metric and evaluated three points: the impact of AD and experimental conditions on SW, the association between SW and delayed recall, and changes in SW across experimental conditions correlated with delayed recall.ResultsSW in the gamma band was significantly lower in patients with AD (z = −2.16, p = 0.031), but the experimental conditions did not exhibit a significant effect in any frequency band. Next, in the AD group, higher scores on delayed recall correlated with diminished SW across delta, alpha, and beta bands in the EO condition. Finally, delayed recall scores significantly predicted relative differences in the SW group in the alpha band (t = −2.98, p = 0.009).DiscussionGiven that network studies could corroborate the results of previous power spectrum studies, our findings contribute to a multifaceted understanding of functional brain networks in AD, emphasizing that the SW properties of these networks change according to disease status, cognitive function, and experimental conditions.
Collapse
Affiliation(s)
- Keigo Yuasa
- Department of Psychiatry and Neurobiology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Tetsu Hirosawa
- Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Daiki Soma
- Department of Psychiatry and Neurobiology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Naoki Furutani
- Department of Psychiatry and Neurobiology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Masafumi Kameya
- Department of Psychiatry and Neurobiology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Masuhiko Sano
- Department of Psychiatry and Neurobiology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Koji Kitamura
- Department of Psychiatry and Neurobiology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Minehisa Ueda
- Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Mitsuru Kikuchi
- Department of Psychiatry and Neurobiology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
26
|
Zhao J, Ikezu TC, Lu W, Macyczko JR, Li Y, Lewis-Tuffin LJ, Martens YA, Ren Y, Zhu Y, Asmann YW, Ertekin-Taner N, Kanekiyo T, Bu G. APOE deficiency impacts neural differentiation and cholesterol biosynthesis in human iPSC-derived cerebral organoids. Stem Cell Res Ther 2023; 14:214. [PMID: 37605285 PMCID: PMC10441762 DOI: 10.1186/s13287-023-03444-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 08/09/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND The apolipoprotein E (APOE) gene is the strongest genetic risk factor for Alzheimer's disease (AD); however, how it modulates brain homeostasis is not clear. The apoE protein is a major lipid carrier in the brain transporting lipids such as cholesterol among different brain cell types. METHODS We generated three-dimensional (3-D) cerebral organoids from human parental iPSC lines and its isogenic APOE-deficient (APOE-/-) iPSC line. To elucidate the cell-type-specific effects of APOE deficiency in the cerebral organoids, we performed scRNA-seq in the parental and APOE-/- cerebral organoids at Day 90. RESULTS We show that APOE deficiency in human iPSC-derived cerebral organoids impacts brain lipid homeostasis by modulating multiple cellular and molecular pathways. Molecular profiling through single-cell RNA sequencing revealed that APOE deficiency leads to changes in cellular composition of isogenic cerebral organoids likely by modulating the eukaryotic initiation factor 2 (EIF2) signaling pathway as these events were alleviated by the treatment of an integrated stress response inhibitor (ISRIB). APOE deletion also leads to activation of the Wnt/β-catenin signaling pathway with concomitant decrease of secreted frizzled-related protein 1 (SFRP1) expression in glia cells. Importantly, the critical role of apoE in cell-type-specific lipid homeostasis was observed upon APOE deletion in cerebral organoids with a specific upregulation of cholesterol biosynthesis in excitatory neurons and excessive lipid accumulation in astrocytes. Relevant to human AD, APOE4 cerebral organoids show altered neurogenesis and cholesterol metabolism compared to those with APOE3. CONCLUSIONS Our work demonstrates critical roles of apoE in brain homeostasis and offers critical insights into the APOE4-related pathogenic mechanisms.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA.
- Center for Regenerative Medicine, Neuroregeneration Laboratory, Mayo Clinic, Jacksonville, FL, 32224, USA.
| | - Tadafumi C Ikezu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Wenyan Lu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Center for Regenerative Medicine, Neuroregeneration Laboratory, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Jesse R Macyczko
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Yonghe Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | | | - Yuka A Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Center for Regenerative Medicine, Neuroregeneration Laboratory, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Yingxue Ren
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Yiyang Zhu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Yan W Asmann
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Department of Neurology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Center for Regenerative Medicine, Neuroregeneration Laboratory, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA.
- Center for Regenerative Medicine, Neuroregeneration Laboratory, Mayo Clinic, Jacksonville, FL, 32224, USA.
| |
Collapse
|
27
|
Abbate C. The Adult Neurogenesis Theory of Alzheimer's Disease. J Alzheimers Dis 2023:JAD221279. [PMID: 37182879 DOI: 10.3233/jad-221279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Alzheimer's disease starts in neural stem cells (NSCs) in the niches of adult neurogenesis. All primary factors responsible for pathological tau hyperphosphorylation are inherent to adult neurogenesis and migration. However, when amyloid pathology is present, it strongly amplifies tau pathogenesis. Indeed, the progressive accumulation of extracellular amyloid-β deposits in the brain triggers a state of chronic inflammation by microglia. Microglial activation has a significant pro-neurogenic effect that fosters the process of adult neurogenesis and supports neuronal migration. Unfortunately, this "reactive" pro-neurogenic activity ultimately perturbs homeostatic equilibrium in the niches of adult neurogenesis by amplifying tau pathogenesis in AD. This scenario involves NSCs in the subgranular zone of the hippocampal dentate gyrus in late-onset AD (LOAD) and NSCs in the ventricular-subventricular zone along the lateral ventricles in early-onset AD (EOAD), including familial AD (FAD). Neuroblasts carrying the initial seed of tau pathology travel throughout the brain via neuronal migration driven by complex signals and convey the disease from the niches of adult neurogenesis to near (LOAD) or distant (EOAD) brain regions. In these locations, or in close proximity, a focus of degeneration begins to develop. Then, tau pathology spreads from the initial foci to large neuronal networks along neural connections through neuron-to-neuron transmission.
Collapse
Affiliation(s)
- Carlo Abbate
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| |
Collapse
|
28
|
Brown J, Camporesi E, Lantero-Rodriguez J, Olsson M, Wang A, Medem B, Zetterberg H, Blennow K, Karikari TK, Wall M, Hill E. Tau in cerebrospinal fluid induces neuronal hyperexcitability and alters hippocampal theta oscillations. Acta Neuropathol Commun 2023; 11:67. [PMID: 37095572 PMCID: PMC10127378 DOI: 10.1186/s40478-023-01562-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/03/2023] [Indexed: 04/26/2023] Open
Abstract
Alzheimer's disease (AD) and other tauopathies are characterized by the aggregation of tau into soluble and insoluble forms (including tangles and neuropil threads). In humans, a fraction of both phosphorylated and non-phosphorylated N-terminal to mid-domain tau species, are secreted into cerebrospinal fluid (CSF). Some of these CSF tau species can be measured as diagnostic and prognostic biomarkers, starting from early stages of disease. While in animal models of AD pathology, soluble tau aggregates have been shown to disrupt neuronal function, it is unclear whether the tau species present in CSF will modulate neural activity. Here, we have developed and applied a novel approach to examine the electrophysiological effects of CSF from patients with a tau-positive biomarker profile. The method involves incubation of acutely-isolated wild-type mouse hippocampal brain slices with small volumes of diluted human CSF, followed by a suite of electrophysiological recording methods to evaluate their effects on neuronal function, from single cells through to the network level. Comparison of the toxicity profiles of the same CSF samples, with and without immuno-depletion for tau, has enabled a pioneering demonstration that CSF-tau potently modulates neuronal function. We demonstrate that CSF-tau mediates an increase in neuronal excitability in single cells. We then observed, at the network level, increased input-output responses and enhanced paired-pulse facilitation as well as an increase in long-term potentiation. Finally, we show that CSF-tau modifies the generation and maintenance of hippocampal theta oscillations, which have important roles in learning and memory and are known to be altered in AD patients. Together, we describe a novel method for screening human CSF-tau to understand functional effects on neuron and network activity, which could have far-reaching benefits in understanding tau pathology, thus allowing for the development of better targeted treatments for tauopathies in the future.
Collapse
Affiliation(s)
- Jessica Brown
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Elena Camporesi
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
| | - Maria Olsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180, Mölndal, Sweden
| | - Alice Wang
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Blanca Medem
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin, Madison, WI, 53792, USA
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180, Mölndal, Sweden
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180, Mölndal, Sweden
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Mark Wall
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Emily Hill
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK.
| |
Collapse
|
29
|
Wander CM, Li YD, Bao H, Asrican B, Luo YJ, Sullivan HA, Chao THH, Zhang WT, Chéry SL, Tart DS, Chen ZK, Shih YYI, Wickersham IR, Cohen TJ, Song J. Compensatory remodeling of a septo-hippocampal GABAergic network in the triple transgenic Alzheimer's mouse model. J Transl Med 2023; 21:258. [PMID: 37061718 PMCID: PMC10105965 DOI: 10.1186/s12967-023-04078-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 03/25/2023] [Indexed: 04/17/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by a progressive loss of memory that cannot be efficiently managed by currently available AD therapeutics. So far, most treatments for AD that have the potential to improve memory target neural circuits to protect their integrity. However, the vulnerable neural circuits and their dynamic remodeling during AD progression remain largely undefined. METHODS Circuit-based approaches, including anterograde and retrograde tracing, slice electrophysiology, and fiber photometry, were used to investigate the dynamic structural and functional remodeling of a GABAergic circuit projected from the medial septum (MS) to the dentate gyrus (DG) in 3xTg-AD mice during AD progression. RESULTS We identified a long-distance GABAergic circuit that couples highly connected MS and DG GABAergic neurons during spatial memory encoding. Furthermore, we found hyperactivity of DG interneurons during early AD, which persisted into late AD stages. Interestingly, MS GABAergic projections developed a series of adaptive strategies to combat DG interneuron hyperactivity. During early-stage AD, MS-DG GABAergic projections exhibit increased inhibitory synaptic strength onto DG interneurons to inhibit their activities. During late-stage AD, MS-DG GABAergic projections form higher anatomical connectivity with DG interneurons and exhibit aberrant outgrowth to increase the inhibition onto DG interneurons. CONCLUSION We report the structural and functional remodeling of the MS-DG GABAergic circuit during disease progression in 3xTg-AD mice. Dynamic MS-DG GABAergic circuit remodeling represents a compensatory mechanism to combat DG interneuron hyperactivity induced by reduced GABA transmission.
Collapse
Affiliation(s)
- Connor M Wander
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Ya-Dong Li
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, 27599, USA.
- Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599, USA.
- Songjiang Research Institute, Songjiang hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201699, China.
| | - Hechen Bao
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Brent Asrican
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Yan-Jia Luo
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Anaesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201699, China
| | - Heather A Sullivan
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Tzu-Hao Harry Chao
- Department of Neurology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Wei-Ting Zhang
- Department of Neurology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Samantha L Chéry
- Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Dalton S Tart
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Ze-Ka Chen
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Yen-Yu Ian Shih
- Department of Neurology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Ian R Wickersham
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Todd J Cohen
- Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Juan Song
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, 27599, USA.
- Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
30
|
Chen XR, Shao Y, Sadowski MJ, on behalf of the Alzheimer’s Disease Neuroimaging Initiative. Interaction between KLOTHO-VS Heterozygosity and APOE ε4 Allele Predicts Rate of Cognitive Decline in Late-Onset Alzheimer's Disease. Genes (Basel) 2023; 14:917. [PMID: 37107675 PMCID: PMC10137709 DOI: 10.3390/genes14040917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/31/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
KLOTHO-VS heterozygosity (KL-VShet+) promotes longevity and protects against cognitive decline in aging. To determine whether KL-VShet+ mitigates Alzheimer's disease (AD) progression, we used longitudinal linear-mixed models to compare the rate of change in multiple cognitive measures in AD patients stratified by APOE ε4 carrier status. We aggregated data on 665 participants (208 KL-VShet-/ε4-, 307 KL-VShet-/ε4+, 66 KL-VShet+/ε4-, and 84 KL-VShet+/ε4+) from two prospective cohorts, the National Alzheimer's Coordinating Center and the Alzheimer's Disease Neuroimaging Initiative. All participants were initially diagnosed with mild cognitive impairment, later developed AD dementia during the study, and had at least three subsequent visits. KL-VShet+ conferred slower cognitive decline in ε4 non-carriers (+0.287 MMSE points/year, p = 0.001; -0.104 CDR-SB points/year, p = 0.026; -0.042 ADCOMS points/year, p < 0.001) but not in ε4 carriers who generally had faster rates of decline than non-carriers. Stratified analyses showed that the protective effect of KL-VShet+ was particularly prominent in male participants, those who were older than the median baseline age of 76 years, or those who had an education level of at least 16 years. For the first time, our study provides evidence that KL-VShet+ status has a protective effect on AD progression and interacts with the ε4 allele.
Collapse
Affiliation(s)
- Xi Richard Chen
- School of Medicine & Dentistry, University of Rochester, Rochester, NY 14642, USA
| | - Yongzhao Shao
- Department of Population Health, NYU Grossman School of Medicine, New York, NY 10016, USA
- Department of Environmental Medicine, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Martin J. Sadowski
- Department of Neurology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY 10016, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | | |
Collapse
|
31
|
Tang Y, Yan Y, Mao J, Ni J, Qing H. The hippocampus associated GABAergic neural network impairment in early-stage of Alzheimer's disease. Ageing Res Rev 2023; 86:101865. [PMID: 36716975 DOI: 10.1016/j.arr.2023.101865] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/13/2023] [Accepted: 01/25/2023] [Indexed: 01/29/2023]
Abstract
Alzheimer's disease (AD) is the commonest neurodegenerative disease with slow progression. Pieces of evidence suggest that the GABAergic system is impaired in the early stage of AD, leading to hippocampal neuron over-activity and further leading to memory and cognitive impairment in patients with AD. However, the precise impairment mechanism of the GABAergic system on the pathogenesis of AD is still unclear. The impairment of neural networks associated with the GABAergic system is tightly associated with AD. Therefore, we describe the roles played by hippocampus-related GABAergic circuits and their impairments in AD neuropathology. In addition, we give our understand on the process from GABAergic circuit impairment to cognitive and memory impairment, since recent studies on astrocyte in AD plays an important role behind cognition dysfunction caused by GABAergic circuit impairment, which helps better understand the GABAergic system and could open up innovative AD therapy.
Collapse
Affiliation(s)
- Yuanhong Tang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Yan Yan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Jian Mao
- Zhengzhou Tobacco Institute of China National Tobacco Company, Zhengzhou 450001, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; Department of Biology, Shenzhen MSU-BIT University, Shenzhen 518172, China.
| |
Collapse
|
32
|
Lee H, Price J, Srivastava DP, Thuret S. In vitro characterization on the role of APOE polymorphism in human hippocampal neurogenesis. Hippocampus 2023; 33:322-346. [PMID: 36709412 PMCID: PMC10947111 DOI: 10.1002/hipo.23502] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/14/2022] [Accepted: 01/11/2023] [Indexed: 01/30/2023]
Abstract
Hippocampal neurogenesis (HN) is considered an important mechanism underlying lifelong brain plasticity, and alterations in this process have been implicated in early Alzheimer's disease progression. APOE polymorphism is the most common genetic risk factor for late-onset Alzheimer's disease where the ε4 genotype is associated with a significantly earlier disease onset compared to the neutral ε3 allele. Recently, APOE has been shown to play an important role in the regulation of HN. However, the time-dependent impact of its polymorphism in humans remains elusive, partially due to the difficulties of studying human HN in vivo. To bridge this gap of knowledge, we used an in vitro cellular model of human HN and performed a time course characterization on isogenic induced pluripotent stem cells with different genotypes of APOE. We found that APOE itself was more highly expressed in ε4 at the stem cell stage, while the divergence of differential gene expression phenotype between ε4 and ε3 became prominent at the neuronal stage of differentiation. This divergence was not associated with the differential capacity to generate dentate gyrus granule cell-like neurons, as its level was comparable between ε4 and ε3. Transcriptomic profiling across different stages of neurogenesis indicated a clear "maturation of functional neurons" phenotype in ε3 neural progenitors and neurons, while genes differentially expressed only in ε4 neurons suggested potential alterations in "metabolism and mitochondrial function." Taken together, our in vitro investigation suggests that APOE ε4 allele can exert a transcriptome-wide effect at the later stages of HN, without altering the overall level of neurogenesis per se.
Collapse
Affiliation(s)
- Hyunah Lee
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| | - Jack Price
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| | - Deepak P. Srivastava
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
- MRC Centre for Neurodevelopmental DisordersKing's College LondonLondonUK
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| |
Collapse
|
33
|
Choi SH, Tanzi RE. Adult neurogenesis in Alzheimer's disease. Hippocampus 2023; 33:307-321. [PMID: 36748337 DOI: 10.1002/hipo.23504] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/08/2023] [Accepted: 01/11/2023] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is the most common form of age-related dementia, characterized by progressive memory loss and cognitive disturbances. The hippocampus, where adult hippocampal neurogenesis (AHN), a relatively novel form of brain plasticity that refers to the birth of new neurons, occurs, is one of the first brain regions to be affected in AD patients. Recent studies showed that AHN persists throughout life in humans, but it drops sharply in AD patients. Next questions to consider would be whether AHN impairment is a contributing factor to learning and memory impairment in AD and whether restoring AHN could ameliorate or delay cognitive dysfunction. Here, we outline and discuss the current knowledge about the state of AHN in AD patients, AHN impairment as a potentially relevant mechanism underlying memory deficits in AD, therapeutic potential of activating AHN in AD, and the mechanisms of AHN impairment in AD.
Collapse
Affiliation(s)
- Se Hoon Choi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
34
|
Rademacher TD, Meuth SG, Wiendl H, Johnen A, Landmeyer NC. Molecular biomarkers and cognitive impairment in multiple sclerosis: State of the field, limitations, and future direction - A systematic review and meta-analysis. Neurosci Biobehav Rev 2023; 146:105035. [PMID: 36608917 DOI: 10.1016/j.neubiorev.2023.105035] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/20/2022] [Accepted: 01/02/2023] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Multiple sclerosis (MS) is associated with cognitive impairment (CI) such as slowed information processing speed (IPS). Currently, no immunocellular or molecular markers have been established in cerebrospinal fluid and serum analysis as surrogate biomarkers with diagnostic or predictive value for the development of CI. This systematic review and meta-analysis aims to sum up the evidence regarding currently discussed markers for CI in MS. METHODS A literature search was conducted on molecular biomarkers of CI in MS, such as neurofilament light chain, chitinases, and vitamin D. RESULTS 5543 publications were screened, of which 77 entered the systematic review. 13 studies were included in the meta-analysis. Neurofilament light chain (CSF: rp = -0.294, p = 0.003; serum: rp = -0.137, p = 0.001) and serum levels of vitamin D (rp = 0.190, p = 0.014) were associated with IPS outcomes. CONCLUSIONS Neurofilament light chain and vitamin D are promising biomarkers to track impairments in IPS in MS. Further longitudinal research is needed to establish the use of molecular biomarkers to monitor cognitive decline.
Collapse
Affiliation(s)
| | - Sven G Meuth
- Department of Neurology, University Hospital Düsseldorf, Germany
| | - Heinz Wiendl
- Department of Neurology, University Hospital Münster, Germany
| | - Andreas Johnen
- Department of Neurology, University Hospital Münster, Germany
| | | |
Collapse
|
35
|
Koutsodendris N, Blumenfeld J, Agrawal A, Traglia M, Grone B, Zilberter M, Yip O, Rao A, Nelson MR, Hao Y, Thomas R, Yoon SY, Arriola P, Huang Y. Neuronal APOE4 removal protects against tau-mediated gliosis, neurodegeneration and myelin deficits. NATURE AGING 2023; 3:275-296. [PMID: 37118426 PMCID: PMC10154214 DOI: 10.1038/s43587-023-00368-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 01/17/2023] [Indexed: 04/30/2023]
Abstract
Apolipoprotein E4 (APOE4) is the strongest known genetic risk factor for late-onset Alzheimer's disease (AD). Conditions of stress or injury induce APOE expression within neurons, but the role of neuronal APOE4 in AD pathogenesis is still unclear. Here we report the characterization of neuronal APOE4 effects on AD-related pathologies in an APOE4-expressing tauopathy mouse model. The selective genetic removal of APOE4 from neurons led to a significant reduction in tau pathology, gliosis, neurodegeneration, neuronal hyperexcitability and myelin deficits. Single-nucleus RNA-sequencing revealed that the removal of neuronal APOE4 greatly diminished neurodegenerative disease-associated subpopulations of neurons, oligodendrocytes, astrocytes and microglia whose accumulation correlated to the severity of tau pathology, neurodegeneration and myelin deficits. Thus, neuronal APOE4 plays a central role in promoting the development of major AD pathologies and its removal can mitigate the progressive cellular and tissue alterations occurring in this model of APOE4-driven tauopathy.
Collapse
Affiliation(s)
- Nicole Koutsodendris
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Jessica Blumenfeld
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Ayushi Agrawal
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA
| | - Michela Traglia
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA
| | - Brian Grone
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Misha Zilberter
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Oscar Yip
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Antara Rao
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Maxine R Nelson
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Yanxia Hao
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Reuben Thomas
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA
| | - Seo Yeon Yoon
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Patrick Arriola
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA.
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA.
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
- Departments of Neurology and Pathology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
36
|
Al-Kuraishy HM, Al-Gareeb AI, Alsayegh AA, Abusudah WF, Almohmadi NH, Eldahshan OA, Ahmed EA, Batiha GES. Insights on benzodiazepines' potential in Alzheimer's disease. Life Sci 2023; 320:121532. [PMID: 36858314 DOI: 10.1016/j.lfs.2023.121532] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/16/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023]
Abstract
Alzheimer's disease (AD) is the most frequent type of dementia characterized by the deposition of amyloid beta (Aβ) plaque and tau-neurofibrillary tangles (TNTs) in the brain. AD is associated with the disturbances of various neurotransmitters including gamma-aminobutyric acid (GABA). Of note, GABA is reduced in AD, and restoration of GABA effect by benzodiazepines (BDZs) may improve AD outcomes. However, BDZs may adversely affect cognitive functions chiefly in elderly AD patients with sleep disorders. Besides, there is a controversy regarding the use of BDZs in AD. Consequently, the objective of the present review was to disclose the possible role of BDZs on the pathogenesis of AD that might be beneficial, neutral, or detrimental effects on AD. Prolonged use of intermediate-acting BDZ lorazepam exerts amnesic effects due to attenuation of synaptic plasticity and impairment of recognition memory. However, BDZs may have a protective effect against the development of AD by reducing tau phosphorylation, neuroinflammation, and progression of AD neuropathology. On the other side, other findings highlighted that extended use of BDZs was not associated with the development of AD. In conclusion, there are controversial points concerning the use of BDZs and the risk for the progression of AD. Thus, preclinical, and clinical studies are essential in this regard.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Bagdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Bagdad, Iraq
| | - Abdulrahman A Alsayegh
- Clinical Nutrition Department Applied Medical Sciences, College Jazan University, Jazan 82817, Saudi Arabia.
| | - Wafaa Fouzi Abusudah
- Clinical Nutrition Department, College of Applied Medical Sciences, Umm Al-Qura University, Makkah 24381, Saudi Arabia.
| | - Najlaa Hamed Almohmadi
- Clinical Nutrition Department, College of Applied Medical Sciences, Umm Al-Qura University, Makkah 24381, Saudi Arabia.
| | - Omayma A Eldahshan
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Eman A Ahmed
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Al Beheira, Egypt.
| |
Collapse
|
37
|
Salta E, Lazarov O, Fitzsimons CP, Tanzi R, Lucassen PJ, Choi SH. Adult hippocampal neurogenesis in Alzheimer's disease: A roadmap to clinical relevance. Cell Stem Cell 2023; 30:120-136. [PMID: 36736288 PMCID: PMC10082636 DOI: 10.1016/j.stem.2023.01.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 02/05/2023]
Abstract
Adult hippocampal neurogenesis (AHN) drops sharply during early stages of Alzheimer's disease (AD), via unknown mechanisms, and correlates with cognitive status in AD patients. Understanding AHN regulation in AD could provide a framework for innovative pharmacological interventions. We here combine molecular, behavioral, and clinical data and critically discuss the multicellular complexity of the AHN niche in relation to AD pathophysiology. We further present a roadmap toward a better understanding of the role of AHN in AD by probing the promises and caveats of the latest technological advancements in the field and addressing the conceptual and methodological challenges ahead.
Collapse
Affiliation(s)
- Evgenia Salta
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
| | - Orly Lazarov
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, 808 S Wood St., Chicago, IL 60612, USA
| | - Carlos P Fitzsimons
- Brain Plasticity group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Rudolph Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, McCance Center for Brain Health, 114 16th Street, Boston, MA 02129, USA.
| | - Paul J Lucassen
- Brain Plasticity group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands; Center for Urban Mental Health, University of Amsterdam, Kruislaan 404, 1098 SM, Amsterdam, The Netherlands.
| | - Se Hoon Choi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, McCance Center for Brain Health, 114 16th Street, Boston, MA 02129, USA.
| |
Collapse
|
38
|
Al-Kuraishy HM, Al-Gareeb AI, Saad HM, Batiha GES. Benzodiazepines in Alzheimer's disease: beneficial or detrimental effects. Inflammopharmacology 2023; 31:221-230. [PMID: 36418599 DOI: 10.1007/s10787-022-01099-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/26/2022]
Abstract
Dementia is considered a clinical syndrome characterized by cognitive dysfunction and memory loss. Alzheimer's disease (AD) is the most common type of dementia. AD is linked with the turbulence of diverse neurotransmitters including gamma-aminobutyric acid (GABA). Notably, GABA in the brain and cerebrospinal fluid was reduced in AD. Thus, allosteric modulation of the GABA effect by benzodiazepines (BDZs) may improve the clinical outcomes of AD patients. Therefore, the objective of the present review was to reveal the possible role of BDZs on the pathogenesis and clinical outcomes in AD patients. Though BDZs may adversely affect cognitive functions mainly in elderly patients, herein it was postulated that BDZs may have beneficial, neutral, or detrimental effects in AD. Taken together, there is strong controversy regarding the use of BDZs and the risk for the development of AD. Therefore, experimental, preclinical and clinical studies are critical to determine the potential protective or detrimental effects of BDZs on AD neuropathology.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Professor in Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Professor in Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Egypt.
| |
Collapse
|
39
|
Lam P, Newland J, Faull RLM, Kwakowsky A. Cation-Chloride Cotransporters KCC2 and NKCC1 as Therapeutic Targets in Neurological and Neuropsychiatric Disorders. Molecules 2023; 28:1344. [PMID: 36771011 PMCID: PMC9920462 DOI: 10.3390/molecules28031344] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023] Open
Abstract
Neurological diseases including Alzheimer's, Huntington's disease, Parkinson's disease, Down syndrome and epilepsy, and neuropsychiatric disorders such as schizophrenia, are conditions that affect not only individuals but societies on a global scale. Current therapies offer a means for small symptomatic relief, but recently there has been increasing demand for therapeutic alternatives. The γ-aminobutyric acid (GABA)ergic signaling system has been investigated for developing new therapies as it has been noted that any dysfunction or changes to this system can contribute to disease progression. Expression of the K-Cl-2 (KCC2) and N-K-C1-1 (NKCC1) cation-chloride cotransporters (CCCs) has recently been linked to the disruption of GABAergic activity by affecting the polarity of GABAA receptor signaling. KCC2 and NKCC1 play a part in multiple neurological and neuropsychiatric disorders, making them a target of interest for potential therapies. This review explores current research suggesting the pathophysiological role and therapeutic importance of KCC2 and NKCC1 in neuropsychiatric and neurological disorders.
Collapse
Affiliation(s)
- Patricia Lam
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Julia Newland
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, 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 1023, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, H91 W5P7 Galway, Ireland
| |
Collapse
|
40
|
Okano H, Takashima K, Takahashi Y, Ojiro R, Tang Q, Ozawa S, Zou X, Koyanagi M, Maronpot RR, Yoshida T, Shibutani M. Progressive disruption of neurodevelopment by mid-gestation exposure to lipopolysaccharides and the ameliorating effect of continuous alpha-glycosyl isoquercitrin treatment. ENVIRONMENTAL TOXICOLOGY 2023; 38:49-69. [PMID: 36125228 DOI: 10.1002/tox.23661] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 06/15/2023]
Abstract
We investigated the effect of lipopolysaccharide (LPS)-induced maternal immune activation used as a model for producing neurodevelopmental disorders on hippocampal neurogenesis and behaviors in rat offspring by exploring the antioxidant effects of alpha-glycosyl isoquercitrin (AGIQ). Pregnant Sprague-Dawley rats were intraperitoneally injected with LPS (50 μg/kg body weight) at gestational days 15 and 16. AGIQ was administered in the diet to dams at 0.5% (w/w) from gestational day 10 until weaning at postnatal day 21 and then to offspring until adulthood at postnatal day 77. During postnatal life, offspring of LPS-injected animals did not show neuroinflammation or oxidative stress in the brain. At weaning, LPS decreased the numbers of type-2b neural progenitor cells (NPCs) and PCNA+ proliferating cells in the subgranular zone, FOS-expressing granule cells, and GAD67+ hilar interneurons in the dentate gyrus. In adulthood, LPS decreased type-1 neural stem cells, type-2a NPCs, and GAD67+ hilar interneurons, and downregulated Dpysl3, Sst, Fos, Mapk1, Mapk3, Grin2a, Grin2b, Bdnf, and Ntrk2. In adults, LPS suppressed locomotor activity in the open field test and suppressed fear memory acquisition and fear extinction learning in the contextual fear conditioning test. These results indicate that mid-gestation LPS injections disrupt programming of normal neurodevelopment resulting in progressive suppression of hippocampal neurogenesis and synaptic plasticity of newborn granule cells by suppressing GABAergic and glutamatergic neurotransmitter signals and BDNF/TrkB signaling to result in adult-stage behavioral deficits. AGIQ ameliorated most aberrations in hippocampal neurogenesis and synaptic plasticity, as well as behavioral deficits. Effective amelioration by continuous AGIQ treatment starting before LPS injections may reflect both anti-inflammatory and anti-oxidative stress effects during gestation and neuroprotective effects of continuous exposure through adulthood.
Collapse
Affiliation(s)
- Hiromu Okano
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Kazumi Takashima
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Yasunori Takahashi
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Ryota Ojiro
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Qian Tang
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Shunsuke Ozawa
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Xinyu Zou
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Mihoko Koyanagi
- Global Scientific and Regulatory Affairs, San-Ei Gen F.F.I. Inc., Osaka, Japan
| | | | - Toshinori Yoshida
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, Tokyo, Japan
| |
Collapse
|
41
|
Steele OG, Stuart AC, Minkley L, Shaw K, Bonnar O, Anderle S, Penn AC, Rusted J, Serpell L, Hall C, King S. A multi-hit hypothesis for an APOE4-dependent pathophysiological state. Eur J Neurosci 2022; 56:5476-5515. [PMID: 35510513 PMCID: PMC9796338 DOI: 10.1111/ejn.15685] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/31/2022] [Accepted: 04/25/2022] [Indexed: 01/01/2023]
Abstract
The APOE gene encoding the Apolipoprotein E protein is the single most significant genetic risk factor for late-onset Alzheimer's disease. The APOE4 genotype confers a significantly increased risk relative to the other two common genotypes APOE3 and APOE2. Intriguingly, APOE4 has been associated with neuropathological and cognitive deficits in the absence of Alzheimer's disease-related amyloid or tau pathology. Here, we review the extensive literature surrounding the impact of APOE genotype on central nervous system dysfunction, focussing on preclinical model systems and comparison of APOE3 and APOE4, given the low global prevalence of APOE2. A multi-hit hypothesis is proposed to explain how APOE4 shifts cerebral physiology towards pathophysiology through interconnected hits. These hits include the following: neurodegeneration, neurovascular dysfunction, neuroinflammation, oxidative stress, endosomal trafficking impairments, lipid and cellular metabolism disruption, impaired calcium homeostasis and altered transcriptional regulation. The hits, individually and in combination, leave the APOE4 brain in a vulnerable state where further cumulative insults will exacerbate degeneration and lead to cognitive deficits in the absence of Alzheimer's disease pathology and also a state in which such pathology may more easily take hold. We conclude that current evidence supports an APOE4 multi-hit hypothesis, which contributes to an APOE4 pathophysiological state. We highlight key areas where further study is required to elucidate the complex interplay between these individual mechanisms and downstream consequences, helping to frame the current landscape of existing APOE-centric literature.
Collapse
Affiliation(s)
| | | | - Lucy Minkley
- School of Life SciencesUniversity of SussexBrightonUK
| | - Kira Shaw
- School of Life SciencesUniversity of SussexBrightonUK
| | - Orla Bonnar
- School of Life SciencesUniversity of SussexBrightonUK
| | | | | | | | | | | | - Sarah King
- School of PsychologyUniversity of SussexBrightonUK
| |
Collapse
|
42
|
Baazaoui N, Iqbal K. Alzheimer's Disease: Challenges and a Therapeutic Opportunity to Treat It with a Neurotrophic Compound. Biomolecules 2022; 12:biom12101409. [PMID: 36291618 PMCID: PMC9599095 DOI: 10.3390/biom12101409] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease with an insidious onset and multifactorial nature. A deficit in neurogenesis and synaptic plasticity are considered the early pathological features associated with neurofibrillary tau and amyloid β pathologies and neuroinflammation. The imbalance of neurotrophic factors with an increase in FGF-2 level and a decrease in brain derived neurotrophic factor (BDNF) and neurotrophin 4 (NT-4) in the hippocampus, frontal cortex and parietal cortex and disruption of the brain micro-environment are other characteristics of AD. Neurotrophic factors are crucial in neuronal differentiation, maturation, and survival. Several attempts to use neurotrophic factors to treat AD were made, but these trials were halted due to their blood-brain barrier (BBB) impermeability, short-half-life, and severe side effects. In the present review we mainly focus on the major etiopathology features of AD and the use of a small neurotrophic and neurogenic peptide mimetic compound; P021 that was discovered in our laboratory and was found to overcome the difficulties faced in the administration of the whole neurotrophic factor proteins. We describe pre-clinical studies on P021 and its potential as a therapeutic drug for AD and related neurodegenerative disorders. Our study is limited because it focuses only on P021 and the relevant literature; a more thorough investigation is required to review studies on various therapeutic approaches and potential drugs that are emerging in the AD field.
Collapse
Affiliation(s)
- Narjes Baazaoui
- Biology Department, College of Sciences and Arts Muhayil Assir, King Khalid University, Abha 61421, Saudi Arabia
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314, USA
- Correspondence: ; Tel.: +1-718-494-5259; Fax: +1-718-494-1080
| |
Collapse
|
43
|
Jeong N, Singer AC. Learning from inhibition: Functional roles of hippocampal CA1 inhibition in spatial learning and memory. Curr Opin Neurobiol 2022; 76:102604. [PMID: 35810533 PMCID: PMC11414469 DOI: 10.1016/j.conb.2022.102604] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/19/2022] [Accepted: 06/07/2022] [Indexed: 11/19/2022]
Abstract
Hippocampal inhibitory interneurons exert a powerful influence on learning and memory. Inhibitory interneurons are known to play a major role in many diseases that affect memory, and to strongly influence brain functions required for memory-related tasks. While previous studies involving genetic, optogenetic, and pharmacological manipulations have shown that hippocampal interneurons play essential roles in spatial and episodic learning and memory, exactly how interneurons affect local circuit computations during spatial navigation is not well understood. Given the significant anatomical, morphological, and functional heterogeneity in hippocampal interneurons, one may suspect cell-type specific roles in circuit computations. Here, we review emerging evidence of CA1 hippocampal interneurons' role in local circuit computations that support spatial learning and memory and discuss open questions about CA1 interneurons in spatial learning.
Collapse
Affiliation(s)
- Nuri Jeong
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA; Neuroscience Graduate Program, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, 30322, USA. https://twitter.com/nuriscientist
| | - Annabelle C Singer
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA.
| |
Collapse
|
44
|
Kim HS, Shin SM, Kim S, Nam Y, Yoo A, Moon M. Relationship between adult subventricular neurogenesis and Alzheimer’s disease: Pathologic roles and therapeutic implications. Front Aging Neurosci 2022; 14:1002281. [PMID: 36185481 PMCID: PMC9518691 DOI: 10.3389/fnagi.2022.1002281] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease that is characterized by irreversible cognitive declines. Senile plaques formed by amyloid-β (Aβ) peptides and neurofibrillary tangles, consisting of hyperphosphorylated tau protein accumulation, are prominent neuropathological features of AD. Impairment of adult neurogenesis is also a well-known pathology in AD. Adult neurogenesis is the process by which neurons are generated from adult neural stem cells. It is closely related to various functions, including cognition, as it occurs throughout life for continuous repair and development of specific neural pathways. Notably, subventricular zone (SVZ) neurogenesis, which occurs in the lateral ventricles, transports neurons to several brain regions such as the olfactory bulb, cerebral cortex, striatum, and hippocampus. These migrating neurons can affect cognitive function and behavior in different neurodegenerative diseases. Despite several studies indicating the importance of adult SVZ neurogenesis in neurodegenerative disorders, the pathological alterations and therapeutic implications of impaired adult neurogenesis in the SVZ in AD have not yet been fully explained. In this review, we summarize recent progress in understanding the alterations in adult SVZ neurogenesis in AD animal models and patients. Moreover, we discuss the potential therapeutic approaches for restoring impaired adult SVZ neurogenesis. Our goal is to impart to readers the importance of adult SVZ neurogenesis in AD and to provide new insights through the discussion of possible therapeutic approaches.
Collapse
Affiliation(s)
- Hyeon Soo Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, South Korea
| | - Seong Min Shin
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, South Korea
| | - Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, South Korea
- Research Institute for Dementia Science, Konyang University, Daejeon, South Korea
| | - Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, South Korea
| | - Anji Yoo
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, South Korea
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, South Korea
- Research Institute for Dementia Science, Konyang University, Daejeon, South Korea
- *Correspondence: Minho Moon,
| |
Collapse
|
45
|
Tarawneh HY, Jayakody DM, Sohrabi HR, Martins RN, Mulders WH. Understanding the Relationship Between Age-Related Hearing Loss and Alzheimer’s Disease: A Narrative Review. J Alzheimers Dis Rep 2022; 6:539-556. [PMID: 36275417 PMCID: PMC9535607 DOI: 10.3233/adr-220035] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/16/2022] [Indexed: 12/02/2022] Open
Abstract
Evidence suggests that hearing loss (HL), even at mild levels, increases the long-term risk of cognitive decline and incident dementia. Hearing loss is one of the modifiable risk factors for dementia, with approximately 4 million of the 50 million cases of dementia worldwide possibly attributed to untreated HL. This paper describes four possible mechanisms that have been suggested for the relationship between age-related hearing loss (ARHL) and Alzheimer’s disease (AD), which is the most common form of dementia. The first mechanism suggests mitochondrial dysfunction and altered signal pathways due to aging as a possible link between ARHL and AD. The second mechanism proposes that sensory degradation in hearing impaired people could explain the relationship between ARHL and AD. The occupation of cognitive resource (third) mechanism indicates that the association between ARHL and AD is a result of increased cognitive processing that is required to compensate for the degraded sensory input. The fourth mechanism is an expansion of the third mechanism, i.e., the function and structure interaction involves both cognitive resource occupation (neural activity) and AD pathology as the link between ARHL and AD. Exploring the specific mechanisms that provide the link between ARHL and AD has the potential to lead to innovative ideas for the diagnosis, prevention, and/or treatment of AD. This paper also provides insight into the current evidence for the use of hearing treatments as a possible treatment/prevention for AD, and if auditory assessments could provide an avenue for early detection of cognitive impairment associated with AD.
Collapse
Affiliation(s)
- Hadeel Y. Tarawneh
- School of Human Sciences, The University of Western Australia, Crawley, WA, Australia
- Ear Science Institute Australia, Subiaco, WA, Australia
| | - Dona M.P. Jayakody
- Ear Science Institute Australia, Subiaco, WA, Australia
- Centre of Ear Science, Medical School, The University of Western Australia, Crawley, WA, Australia
| | - Hamid R. Sohrabi
- Centre for Healthy Ageing, College of Science, Health, Engineering and Education, Murdoch University, WA, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, NSW, Australia
| | - Ralph N. Martins
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, NSW, Australia
| | | |
Collapse
|
46
|
Shang Q, Chen G, Zhang P, Zhao W, Chen H, Yu D, Yu F, Liu H, Zhang X, He J, Yu X, Zhang Z, Tan R, Wu Z, Tang J, Liang D, Shen G, Jiang X, Ren H. Myristic acid alleviates hippocampal aging correlated with GABAergic signaling. Front Nutr 2022; 9:907526. [PMID: 36159502 PMCID: PMC9493098 DOI: 10.3389/fnut.2022.907526] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
Previous studies have shown that myristic acid (MA), a saturated fatty acid, could promote the proliferation and differentiation of neural stem cells in vitro. However, the effect of MA on hippocampal neurons aging has not been reported in vivo. Here we employed 22-month-old naturally aged C57BL/6 mice to evaluate the effect and mechanism of MA on hippocampal aging. First, we examined a decreased exploration and spatial memory ability in aging mice using the open field test and Morris water maze. Consistently, aging mice showed degenerative hippocampal histomorphology by H&E and Nissl staining. In terms of mechanism, imbalance of GABRB2 and GABRA2 expression in aging mice might be involved in hippocampus aging by mRNA high throughput sequencing (mRNA-seq) and immunohistochemistry (IHC) validation. Then, we revealed that MA alleviated the damage of exploration and spatial memory ability and ameliorated degeneration and aging of hippocampal neurons. Meanwhile, MA downregulated GABRB2 and upregulated GABRA2 expression, indicating MA might alleviate hippocampal aging correlated with GABAergic signaling. In conclusion, our findings revealed MA alleviated hippocampal aging correlated with GABAergic signaling, which might provide insight into the treatment of aging-associated diseases.
Collapse
Affiliation(s)
- Qi Shang
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guifeng Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peng Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenhua Zhao
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Honglin Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Die Yu
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fuyong Yu
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huiwen Liu
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuelai Zhang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiahui He
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiang Yu
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhida Zhang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Riwei Tan
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zixian Wu
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingjing Tang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - De Liang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Gengyang Shen
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Nanshan Hospital, The First Affiliated Hospital of Guangzhou University of Chinese Medicine (Shenzhen Nanshan Hospital of Chinese Medicine), Guangzhou, China
- Gengyang Shen
| | - Xiaobing Jiang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Xiaobing Jiang
| | - Hui Ren
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Hui Ren
| |
Collapse
|
47
|
Gutierrez BA, Limon A. Synaptic Disruption by Soluble Oligomers in Patients with Alzheimer's and Parkinson's Disease. Biomedicines 2022; 10:1743. [PMID: 35885050 PMCID: PMC9313353 DOI: 10.3390/biomedicines10071743] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 01/10/2023] Open
Abstract
Neurodegenerative diseases are the result of progressive dysfunction of the neuronal activity and subsequent neuronal death. Currently, the most prevalent neurodegenerative diseases are by far Alzheimer's (AD) and Parkinson's (PD) disease, affecting millions of people worldwide. Although amyloid plaques and neurofibrillary tangles are the neuropathological hallmarks for AD and Lewy bodies (LB) are the hallmark for PD, current evidence strongly suggests that oligomers seeding the neuropathological hallmarks are more toxic and disease-relevant in both pathologies. The presence of small soluble oligomers is the common bond between AD and PD: amyloid β oligomers (AβOs) and Tau oligomers (TauOs) in AD and α-synuclein oligomers (αSynOs) in PD. Such oligomers appear to be particularly increased during the early pathological stages, targeting synapses at vulnerable brain regions leading to synaptic plasticity disruption, synapse loss, inflammation, excitation to inhibition imbalance and cognitive impairment. Absence of TauOs at synapses in individuals with strong AD disease pathology but preserved cognition suggests that mechanisms of resilience may be dependent on the interactions between soluble oligomers and their synaptic targets. In this review, we will discuss the current knowledge about the interactions between soluble oligomers and synaptic dysfunction in patients diagnosed with AD and PD, how it affects excitatory and inhibitory synaptic transmission, and the potential mechanisms of synaptic resilience in humans.
Collapse
Affiliation(s)
| | - Agenor Limon
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, School of Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA;
| |
Collapse
|
48
|
Targa Dias Anastacio H, Matosin N, Ooi L. Neuronal hyperexcitability in Alzheimer's disease: what are the drivers behind this aberrant phenotype? Transl Psychiatry 2022; 12:257. [PMID: 35732622 PMCID: PMC9217953 DOI: 10.1038/s41398-022-02024-7] [Citation(s) in RCA: 160] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/01/2022] [Accepted: 06/08/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder leading to loss of cognitive abilities and ultimately, death. With no cure available, limited treatments mostly focus on symptom management. Identifying early changes in the disease course may provide new therapeutic targets to halt or reverse disease progression. Clinical studies have shown that cortical and hippocampal hyperactivity are a feature shared by patients in the early stages of disease, progressing to hypoactivity during later stages of neurodegeneration. The exact mechanisms causing neuronal excitability changes are not fully characterized; however, animal and cell models have provided insights into some of the factors involved in this phenotype. In this review, we summarize the evidence for neuronal excitability changes over the course of AD onset and progression and the molecular mechanisms underpinning these differences. Specifically, we discuss contributors to aberrant neuronal excitability, including abnormal levels of intracellular Ca2+ and glutamate, pathological amyloid β (Aβ) and tau, genetic risk factors, including APOE, and impaired inhibitory interneuron and glial function. In light of recent research indicating hyperexcitability could be a predictive marker of cognitive dysfunction, we further argue that the hyperexcitability phenotype could be leveraged to improve the diagnosis and treatment of AD, and present potential targets for future AD treatment development.
Collapse
Affiliation(s)
- Helena Targa Dias Anastacio
- grid.510958.0Illawarra Health and Medical Research Institute, Wollongong, NSW 2522 Australia ,grid.1007.60000 0004 0486 528XMolecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522 Australia
| | - Natalie Matosin
- grid.510958.0Illawarra Health and Medical Research Institute, Wollongong, NSW 2522 Australia ,grid.1007.60000 0004 0486 528XMolecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522 Australia
| | - Lezanne Ooi
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia. .,Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2522, Australia.
| |
Collapse
|
49
|
Capsoni S, Arisi I, Malerba F, D’Onofrio M, Cattaneo A, Cherubini E. Targeting the Cation-Chloride Co-Transporter NKCC1 to Re-Establish GABAergic Inhibition and an Appropriate Excitatory/Inhibitory Balance in Selective Neuronal Circuits: A Novel Approach for the Treatment of Alzheimer's Disease. Brain Sci 2022; 12:783. [PMID: 35741668 PMCID: PMC9221351 DOI: 10.3390/brainsci12060783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 01/27/2023] Open
Abstract
GABA, the main inhibitory neurotransmitter in the adult brain, depolarizes and excites immature neurons because of an initially higher intracellular chloride concentration [Cl-]i due to the delayed expression of the chloride exporter KCC2 at birth. Depolarization-induced calcium rise via NMDA receptors and voltage-dependent calcium channels is instrumental in shaping neuronal circuits and in controlling the excitatory (E)/inhibitory (I) balance in selective brain areas. An E/I imbalance accounts for cognitive impairment observed in several neuropsychiatric disorders. The aim of this review is to summarize recent data on the mechanisms by which alterations of GABAergic signaling alter the E/I balance in cortical and hippocampal neurons in Alzheimer's disease (AD) and the role of cation-chloride co-transporters in this process. In particular, we discuss the NGF and AD relationship and how mice engineered to express recombinant neutralizing anti-NGF antibodies (AD11 mice), which develop a neurodegenerative pathology reminiscent of that observed in AD patients, exhibit a depolarizing action of GABA due to KCC2 impairment. Treating AD and other forms of dementia with bumetanide, a selective KCC2 antagonist, contributes to re-establishing a proper E/I balance in selective brain areas, leading to amelioration of AD symptoms and the slowing down of disease progression.
Collapse
Affiliation(s)
- Simona Capsoni
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy;
- Section of Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| | - Ivan Arisi
- Fondazione European Brain Research Institute (EBRI) Rita Levi-Montalcini, 00161 Rome, Italy; (I.A.); (F.M.); (M.D.)
| | - Francesca Malerba
- Fondazione European Brain Research Institute (EBRI) Rita Levi-Montalcini, 00161 Rome, Italy; (I.A.); (F.M.); (M.D.)
| | - Mara D’Onofrio
- Fondazione European Brain Research Institute (EBRI) Rita Levi-Montalcini, 00161 Rome, Italy; (I.A.); (F.M.); (M.D.)
| | - Antonino Cattaneo
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy;
- Fondazione European Brain Research Institute (EBRI) Rita Levi-Montalcini, 00161 Rome, Italy; (I.A.); (F.M.); (M.D.)
| | - Enrico Cherubini
- Fondazione European Brain Research Institute (EBRI) Rita Levi-Montalcini, 00161 Rome, Italy; (I.A.); (F.M.); (M.D.)
| |
Collapse
|
50
|
Culig L, Chu X, Bohr VA. Neurogenesis in aging and age-related neurodegenerative diseases. Ageing Res Rev 2022; 78:101636. [PMID: 35490966 PMCID: PMC9168971 DOI: 10.1016/j.arr.2022.101636] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/14/2022] [Accepted: 04/25/2022] [Indexed: 12/11/2022]
Abstract
Adult neurogenesis, the process by which neurons are generated in certain areas of the adult brain, declines in an age-dependent manner and is one potential target for extending cognitive healthspan. Aging is a major risk factor for neurodegenerative diseases and, as lifespans are increasing, these health challenges are becoming more prevalent. An age-associated loss in neural stem cell number and/or activity could cause this decline in brain function, so interventions that reverse aging in stem cells might increase the human cognitive healthspan. In this review, we describe the involvement of adult neurogenesis in neurodegenerative diseases and address the molecular mechanistic aspects of neurogenesis that involve some of the key aggregation-prone proteins in the brain (i.e., tau, Aβ, α-synuclein, …). We summarize the research pertaining to interventions that increase neurogenesis and regulate known targets in aging research, such as mTOR and sirtuins. Lastly, we share our outlook on restoring the levels of neurogenesis to physiological levels in elderly individuals and those with neurodegeneration. We suggest that modulating neurogenesis represents a potential target for interventions that could help in the fight against neurodegeneration and cognitive decline.
Collapse
Affiliation(s)
- Luka Culig
- Section on DNA Repair, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Xixia Chu
- Section on DNA Repair, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Vilhelm A Bohr
- Section on DNA Repair, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|