1
|
Mohd Murshid N, Mohd Sahardi NFN, Makpol S. Advancing Alzheimer's Disease Modelling by Developing a Refined Biomimetic Brain Microenvironment for Facilitating High-Throughput Screening of Pharmacological Treatment Strategies. Int J Mol Sci 2024; 26:241. [PMID: 39796097 PMCID: PMC11719782 DOI: 10.3390/ijms26010241] [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: 10/18/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 01/13/2025] Open
Abstract
Alzheimer's disease (AD) poses a significant worldwide health challenge, requiring novel approaches for improved models and treatment development. This comprehensive review emphasises the systematic development and improvement of a biomimetic brain environment to address the shortcomings of existing AD models and enhance the efficiency of screening potential drug treatments. We identify drawbacks in traditional models and emphasise the necessity for more physiologically accurate systems through an in-depth analysis of current literature. This review aims to study the development of an advanced AD model that accurately replicates key AD pathophysiological aspects using cutting-edge biomaterials and microenvironment design. Incorporating biomolecular elements like Tau proteins and beta-amyloid (Aβ) plaques improve the accuracy of illustrating disease mechanisms. The expected results involve creating a solid foundation for high-throughput screening with enhanced scalability, translational significance, and the possibility of speeding up drug discovery. Thus, this review fills the gaps in AD modelling and shows potential for creating precise and efficient drug treatments for AD.
Collapse
Affiliation(s)
- Nuraqila Mohd Murshid
- Department of Biochemistry, Faculty of Medicine, Level 17 Preclinical Building, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Nur Fatin Nabilah Mohd Sahardi
- Secretariat of Research and Innovation, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, Level 17 Preclinical Building, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
2
|
Collins HM, Greenfield S. Rodent Models of Alzheimer's Disease: Past Misconceptions and Future Prospects. Int J Mol Sci 2024; 25:6222. [PMID: 38892408 PMCID: PMC11172947 DOI: 10.3390/ijms25116222] [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: 04/03/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease with no effective treatments, not least due to the lack of authentic animal models. Typically, rodent models recapitulate the effects but not causes of AD, such as cholinergic neuron loss: lesioning of cholinergic neurons mimics the cognitive decline reminiscent of AD but not its neuropathology. Alternative models rely on the overexpression of genes associated with familial AD, such as amyloid precursor protein, or have genetically amplified expression of mutant tau. Yet transgenic rodent models poorly replicate the neuropathogenesis and protein overexpression patterns of sporadic AD. Seeding rodents with amyloid or tau facilitates the formation of these pathologies but cannot account for their initial accumulation. Intracerebral infusion of proinflammatory agents offer an alternative model, but these fail to replicate the cause of AD. A novel model is therefore needed, perhaps similar to those used for Parkinson's disease, namely adult wildtype rodents with neuron-specific (dopaminergic) lesions within the same vulnerable brainstem nuclei, 'the isodendritic core', which are the first to degenerate in AD. Site-selective targeting of these nuclei in adult rodents may recapitulate the initial neurodegenerative processes in AD to faithfully mimic its pathogenesis and progression, ultimately leading to presymptomatic biomarkers and preventative therapies.
Collapse
Affiliation(s)
- Helen M. Collins
- Neuro-Bio Ltd., Building F5 The Culham Campus, Abingdon OX14 3DB, UK;
| | | |
Collapse
|
3
|
Lee MK, Chen G. Loss of Cholinergic and Monoaminergic Afferents in APPswe/PS1ΔE9 Transgenic Mouse Model of Cerebral Amyloidosis Preferentially Occurs Near Amyloid Plaques. Int J Mol Sci 2024; 25:5004. [PMID: 38732223 PMCID: PMC11084680 DOI: 10.3390/ijms25095004] [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: 03/28/2024] [Revised: 04/22/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by a loss of neurons in the cortex and subcortical regions. Previously, we showed that the progressive degeneration of subcortical monoaminergic (MAergic) neurons seen in human AD is recapitulated in the APPswe/PS1ΔE9 (APP/PS) transgenic mouse model. Because degeneration of cholinergic (Ach) neurons is also a prominent feature of AD, we examined the integrity of the Ach system in the APP/PS model. The overall density of Ach fibers is reduced in APP/PS1 mice at 12 and 18 months of age but not at 4 months of age. Analysis of basal forebrain Ach neurons shows no loss of Ach neurons in the APP/PS model. Thus, since MAergic systems show overt cell loss at 18 months of age, the Ach system is less vulnerable to neurodegeneration in the APP/PS1 model. We also examined whether the proximity to Aβ deposition affected the degeneration of Ach and 5-HT afferents. We found that the areas closer to the edges of compact Aβ deposits exhibit a more severe loss of afferents than the areas that are more distal to Aβ deposits. Collectively, the results indicate that the APP/PS model recapitulates the degeneration of multiple subcortical neurotransmitter systems, including the Ach system. In addition, the results indicate that Aβ deposits cause global as well as local toxicity to subcortical afferents.
Collapse
Affiliation(s)
- Michael K. Lee
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Gang Chen
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Personalized Medicine in Brain Disorders, The Guangdong-Hongkong-Macao Joint Laboratory of TCM on Brain-Peripheral Homeostasis and Comprehensive Health, School of Chinese Medicine, Jinan University, Guangzhou 510632, China
| |
Collapse
|
4
|
Qiu T, Hong H, Zeng Q, Luo X, Wang X, Xu X, Xie F, Li X, Li K, Huang P, Dai S, Zhang M. Degeneration of cholinergic white matter pathways and nucleus basalis of Meynert in individuals with objective subtle cognitive impairment. Neurobiol Aging 2023; 132:198-208. [PMID: 37852044 DOI: 10.1016/j.neurobiolaging.2023.09.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/20/2023]
Abstract
We evaluated alterations in the nucleus basalis of Meynert (NBM) volume and integrity of cholinergic white matter pathways in objective subtle cognitive impairment (Obj-SCI) individuals. NBM segmentation and cholinergic pathways tracking were conducted at baseline, 12-, 24-, and 48-month follow-ups in 41 Obj-SCI individuals and 61 healthy controls (HC). The baseline and 4-year rate of change in NBM volume and cholinergic pathways mean diffusivity were compared. Associations between cholinergic index changes and pathological processes and cognitive performance were evaluated. After controlling for age, sex, APOE genotype, and total intracranial volume, Obj-SCI individuals exhibited reduced NBM volume and increased medial pathway mean diffusivity compared to HC at baseline. Furthermore, amyloid-positive Obj-SCI individuals exhibited a steeper longitudinal decline in NBM volume than HC. Additionally, decreases in NBM volume and cholinergic pathways integrity were associated with amyloid and vascular pathologies and cognitive decline. Overall, degeneration of the cholinergic system plays an important role in cognitive impairment during the preclinical stage of Alzheimer's disease, which may provide a significant target for early therapeutic interventions.
Collapse
Affiliation(s)
- Tiantian Qiu
- Department of Radiology, Linyi People's Hospital, Linyi, China
| | - Hui Hong
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Qingze Zeng
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Luo
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaohan Wang
- Clinical Laboratory, Linyi Central Hospital, Linyi, China
| | - Xiaopei Xu
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Xie
- Department of Equipment and Medical Engineering, Linyi People's Hospital, Linyi, China
| | - Xiaodong Li
- Department of Radiology, Linyi People's Hospital, Linyi, China
| | - Kaicheng Li
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Peiyu Huang
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Shouping Dai
- Department of Radiology, Linyi People's Hospital, Linyi, China.
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
5
|
Rozalem Aranha M, Iulita MF, Montal V, Pegueroles J, Bejanin A, Vaqué-Alcázar L, Grothe MJ, Carmona-Iragui M, Videla L, Benejam B, Arranz J, Padilla C, Valldeneu S, Barroeta I, Altuna M, Fernández S, Ribas L, Valle-Tamayo N, Alcolea D, González-Ortiz S, Bargalló N, Zetterberg H, Blennow K, Blesa R, Wisniewski T, Busciglio J, Cuello AC, Lleó A, Fortea J. Basal forebrain atrophy along the Alzheimer's disease continuum in adults with Down syndrome. Alzheimers Dement 2023; 19:4817-4827. [PMID: 37021589 DOI: 10.1002/alz.12999] [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: 10/03/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Basal forebrain (BF) degeneration occurs in Down syndrome (DS)-associated Alzheimer's disease (AD). However, the dynamics of BF atrophy with age and disease progression, its impact on cognition, and its relationship with AD biomarkers have not been studied in DS. METHODS We included 234 adults with DS (150 asymptomatic, 38 prodromal AD, and 46 AD dementia) and 147 euploid controls. BF volumes were extracted from T-weighted magnetic resonance images using a stereotactic atlas in SPM12. We assessed BF volume changes with age and along the clinical AD continuum and their relationship to cognitive performance, cerebrospinal fluid (CSF) and plasma amyloid/tau/neurodegeneration biomarkers, and hippocampal volume. RESULTS In DS, BF volumes decreased with age and along the clinical AD continuum and significantly correlated with amyloid, tau, and neurofilament light chain changes in CSF and plasma, hippocampal volume, and cognitive performance. DISCUSSION BF atrophy is a potentially valuable neuroimaging biomarker of AD-related cholinergic neurodegeneration in DS.
Collapse
Affiliation(s)
- Mateus Rozalem Aranha
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Maria Florencia Iulita
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Victor Montal
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jordi Pegueroles
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Alexandre Bejanin
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Lídia Vaqué-Alcázar
- Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Department of Medicine, Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Michel J Grothe
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Maria Carmona-Iragui
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Laura Videla
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Bessy Benejam
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Javier Arranz
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Concepción Padilla
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Sílvia Valldeneu
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Isabel Barroeta
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Miren Altuna
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Susana Fernández
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Laia Ribas
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Natalia Valle-Tamayo
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Sofía González-Ortiz
- Hospital del Mar - Parc de Salut Mar, Barcelona, Spain
- Neuroradiology Section, Radiology Department, Diagnostic Image Center, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Núria Bargalló
- Neuroradiology Section, Radiology Department, Diagnostic Image Center, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
- Magnetic Resonance Image Core Facility (IDIBAPS), Barcelona, Spain
| | - Henrik Zetterberg
- Queen Square Institute of Neurology, University College London, London, UK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- UK Dementia Research Institute, University College London, London, UK
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Hong Kong Center for Neurodegenerative Diseases, China, Hong Kong
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Rafael Blesa
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Thomas Wisniewski
- Departments of Neurology, Pathology and Psychiatry and Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, New York, USA
| | - Jorge Busciglio
- Department of Neurobiology & Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), Center for the Neurobiology of Learning and Memory, University of California at Irvine, Irvine, California, USA
| | - A Claudio Cuello
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
- Department of Pharmacology, Oxford University, Oxford, UK
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| |
Collapse
|
6
|
Daamen M, Scheef L, Li S, Grothe MJ, Gaertner FC, Buchert R, Buerger K, Dobisch L, Drzezga A, Essler M, Ewers M, Fliessbach K, Herrera Melendez AL, Hetzer S, Janowitz D, Kilimann I, Krause BJ, Lange C, Laske C, Munk MH, Peters O, Priller J, Ramirez A, Reimold M, Rominger A, Rostamzadeh A, Roeske S, Roy N, Scheffler K, Schneider A, Spottke A, Spruth EJ, Teipel SJ, Wagner M, Düzel E, Jessen F, Boecker H. Cortical Amyloid Burden Relates to Basal Forebrain Volume in Subjective Cognitive Decline. J Alzheimers Dis 2023; 95:1013-1028. [PMID: 37638433 DOI: 10.3233/jad-230141] [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] [Indexed: 08/29/2023]
Abstract
BACKGROUND Atrophy of cholinergic basal forebrain (BF) nuclei is a frequent finding in magnetic resonance imaging (MRI) volumetry studies that examined patients with prodromal or clinical Alzheimer's disease (AD), but less clear for individuals in earlier stages of the clinical AD continuum. OBJECTIVE To examine BF volume reductions in subjective cognitive decline (SCD) participants with AD pathologic changes. METHODS The present study compared MRI-based BF volume measurements in age- and sex-matched samples of N = 24 amyloid-positive and N = 24 amyloid-negative SCD individuals, based on binary visual ratings of Florbetaben positron emission tomography (PET) measurements. Additionally, we assessed associations of BF volume with cortical amyloid burden, based on semiquantitative Centiloid (CL) analyses. RESULTS Group differences approached significance for BF total volume (p = 0.061) and the Ch4 subregion (p = 0.059) only, showing the expected relative volume reductions for the amyloid-positive subgroup. There were also significant inverse correlations between BF volumes and CL values, which again were most robust for BF total volume and the Ch4 subregion. CONCLUSIONS The results are consistent with the hypothesis that amyloid-positive SCD individuals, which are considered to represent a transitional stage on the clinical AD continuum, already show incipient alterations of BF integrity. The negative association with a continuous measure of cortical amyloid burden also suggests that this may reflect an incremental process. Yet, further research is needed to evaluate whether BF changes already emerge at "grey zone" levels of amyloid accumulation, before amyloidosis is reliably detected by PET visual readings.
Collapse
Affiliation(s)
- Marcel Daamen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Lukas Scheef
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department for Diagnostic and Interventional Radiology, University Hospital Bonn, Bonn, Germany
- RheinAhrCampus, University of Applied Sciences Koblenz, Remagen, Germany
| | - Shumei Li
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Michel J Grothe
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | | | - Ralph Buchert
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Buerger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian University Munich, Munich, Germany
| | - Laura Dobisch
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Alexander Drzezga
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-2), Molecular Organization of the Brain, Forschungszentrum Jülich, Jülich, Germany
| | - Markus Essler
- Department of Nuclear Medicine, University Hospital Bonn, Bonn, Germany
| | - Michael Ewers
- Institute for Clinical Radiology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Klaus Fliessbach
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
| | - Ana Lucia Herrera Melendez
- Institute of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stefan Hetzer
- Berlin Center of Advanced Neuroimaging, Charité -Universitätsmedizin Berlin, Berlin, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian University Munich, Munich, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Bernd Joachim Krause
- Department of Nuclear Medicine, Rostock University Medical Centre, Rostock, Germany
| | - Catharina Lange
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE), Tuebingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Matthias H Munk
- German Center for Neurodegenerative Diseases (DZNE), Tuebingen, Germany
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Oliver Peters
- Institute of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany
- University of Edinburgh and UK Dementia Research Institute, Edinburgh, UK
| | - Alfredo Ramirez
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Department of Psychiatry and Psychotherapy, Division of Neurogenetics and Molecular Psychiatry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Medical Faculty, Cologne, Germany
- Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, San Antonio, TX, USA
| | - Matthias Reimold
- Department of Nuclear Medicine and Clinical Molecular Imaging, Eberhard-Karls-University, Tübingen, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, Ludwig-Maximilian-University Munich, Munich, Germany
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ayda Rostamzadeh
- Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Sandra Roeske
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Nina Roy
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Klaus Scheffler
- Department for Biomedical Magnetic Resonance, University of Tübingen, Tübingen, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Eike Jakob Spruth
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stefan J Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Henning Boecker
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department for Diagnostic and Interventional Radiology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
7
|
Sanchez-Varo R, Mejias-Ortega M, Fernandez-Valenzuela JJ, Nuñez-Diaz C, Caceres-Palomo L, Vegas-Gomez L, Sanchez-Mejias E, Trujillo-Estrada L, Garcia-Leon JA, Moreno-Gonzalez I, Vizuete M, Vitorica J, Baglietto-Vargas D, Gutierrez A. Transgenic Mouse Models of Alzheimer's Disease: An Integrative Analysis. Int J Mol Sci 2022; 23:5404. [PMID: 35628216 PMCID: PMC9142061 DOI: 10.3390/ijms23105404] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/10/2022] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) constitutes the most prominent form of dementia among elderly individuals worldwide. Disease modeling using murine transgenic mice was first initiated thanks to the discovery of heritable mutations in amyloid precursor protein (APP) and presenilins (PS) genes. However, due to the repeated failure of translational applications from animal models to human patients, along with the recent advances in genetic susceptibility and our current understanding on disease biology, these models have evolved over time in an attempt to better reproduce the complexity of this devastating disease and improve their applicability. In this review, we provide a comprehensive overview about the major pathological elements of human AD (plaques, tauopathy, synaptic damage, neuronal death, neuroinflammation and glial dysfunction), discussing the knowledge that available mouse models have provided about the mechanisms underlying human disease. Moreover, we highlight the pros and cons of current models, and the revolution offered by the concomitant use of transgenic mice and omics technologies that may lead to a more rapid improvement of the present modeling battery.
Collapse
Affiliation(s)
- Raquel Sanchez-Varo
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
- Departamento Fisiologia Humana, Histologia Humana, Anatomia Patologica y Educacion Fisica y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071 Malaga, Spain
| | - Marina Mejias-Ortega
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Juan Jose Fernandez-Valenzuela
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Cristina Nuñez-Diaz
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Laura Caceres-Palomo
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Laura Vegas-Gomez
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Elisabeth Sanchez-Mejias
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Laura Trujillo-Estrada
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Juan Antonio Garcia-Leon
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Ines Moreno-Gonzalez
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Marisa Vizuete
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
- Departamento Bioquimica y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, Instituto de Biomedicina de Sevilla (IBIS)-Hospital Universitario Virgen del Rocio/CSIC, 41012 Seville, Spain
| | - Javier Vitorica
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
- Departamento Bioquimica y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, Instituto de Biomedicina de Sevilla (IBIS)-Hospital Universitario Virgen del Rocio/CSIC, 41012 Seville, Spain
| | - David Baglietto-Vargas
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Antonia Gutierrez
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| |
Collapse
|
8
|
Pons V, Rivest S. Targeting Systemic Innate Immune Cells as a Therapeutic Avenue for Alzheimer Disease. Pharmacol Rev 2022; 74:1-17. [PMID: 34987086 DOI: 10.1124/pharmrev.121.000400] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer disease (AD) is the first progressive neurodegenerative disease worldwide, and the disease is characterized by an accumulation of amyloid in the brain and neurovasculature that triggers cognitive decline and neuroinflammation. The innate immune system has a preponderant role in AD. The last decade, scientists focused their efforts on therapies aiming to modulate innate immunity. The latter is of great interest, since they participate to the inflammation and phagocytose the amyloid in the brain and blood vessels. We and others have developed pharmacological approaches to stimulate these cells using various ligands. These include toll-like receptor 4, macrophage colony stimulating factor, and more recently nucleotide-binding oligomerization domain-containing 2 receptors. This review will discuss the great potential to take advantage of the innate immune system to fight naturally against amyloid β accumulation and prevent its detrimental consequence on brain functions and its vascular system. SIGNIFICANCE STATEMENT: The focus on amyloid β removal from the perivascular space rather than targeting CNS plaque formation and clearance represents a new direction with a great potential. Small molecules able to act at the level of peripheral immunity would constitute a novel approach for tackling aberrant central nervous system biology, one of which we believe would have the potential of generating a lot of interest.
Collapse
Affiliation(s)
- Vincent Pons
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Boul., Québec City, QC G1V 4G2, Canada
| | - Serge Rivest
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Boul., Québec City, QC G1V 4G2, Canada
| |
Collapse
|
9
|
Cantero JL, Atienza M, Lage C, Zaborszky L, Vilaplana E, Lopez-Garcia S, Pozueta A, Rodriguez-Rodriguez E, Blesa R, Alcolea D, Lleo A, Sanchez-Juan P, Fortea J. Atrophy of Basal Forebrain Initiates with Tau Pathology in Individuals at Risk for Alzheimer's Disease. Cereb Cortex 2021; 30:2083-2098. [PMID: 31799623 DOI: 10.1093/cercor/bhz224] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/30/2019] [Accepted: 08/28/2019] [Indexed: 12/12/2022] Open
Abstract
Evidence suggests that the basal forebrain (BF) cholinergic system degenerates early in the course of Alzheimer's disease (AD), likely due to the vulnerability of BF cholinergic neurons to tau pathology. However, it remains unclear whether the presence of tauopathy is the only requirement for initiating the BF degeneration in asymptomatic subjects at risk for AD (AR-AD), and how BF structural deficits evolve from normal aging to preclinical and prodromal AD. Here, we provide human in vivo magnetic resonance imaging evidence supporting that abnormal cerebrospinal fluid levels of phosphorylated tau (T+) are selectively associated with bilateral volume loss of the nucleus basalis of Meynert (nbM, Ch4) in AR-AD individuals. Spreading of atrophy to medial septum and vertical limb of diagonal band Broca (Ch1-Ch2) occurred in both preclinical and prodromal AD. With the exception of A+, all groups revealed significant correlations between volume reduction of BF cholinergic compartments and atrophy of their innervated regions. Overall, these results support the central role played by tauopathy in instigating the nbM degeneration in AR-AD individuals and the necessary coexistence of both AD proteinopathies for spreading damage to larger BF territories, thus affecting the core of the BF cholinergic projection system.
Collapse
Affiliation(s)
- Jose L Cantero
- Laboratory of Functional Neuroscience, Pablo de Olavide University, 41013 Seville, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
| | - Mercedes Atienza
- Laboratory of Functional Neuroscience, Pablo de Olavide University, 41013 Seville, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
| | - Carmen Lage
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Service of Neurology, IDIVAL, University Hospital Marques de Valdecilla, University of Cantabria, 39008 Santander, Spain
| | - Laszlo Zaborszky
- Center for Molecular and Behavioral Neuroscience, Rutgers, The State University of New Jersey, Newark, 07102 NJ, USA
| | - Eduard Vilaplana
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Department of Neurology, Institut d'Investigacions Biomediques Sant Pau-Hospital Santa Creu i Sant Pau, Universitat Autonoma de Barcelona, 08025 Barcelona, Spain
| | - Sara Lopez-Garcia
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Service of Neurology, IDIVAL, University Hospital Marques de Valdecilla, University of Cantabria, 39008 Santander, Spain
| | - Ana Pozueta
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Service of Neurology, IDIVAL, University Hospital Marques de Valdecilla, University of Cantabria, 39008 Santander, Spain
| | - Eloy Rodriguez-Rodriguez
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Service of Neurology, IDIVAL, University Hospital Marques de Valdecilla, University of Cantabria, 39008 Santander, Spain
| | - Rafael Blesa
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Department of Neurology, Institut d'Investigacions Biomediques Sant Pau-Hospital Santa Creu i Sant Pau, Universitat Autonoma de Barcelona, 08025 Barcelona, Spain
| | - Daniel Alcolea
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Department of Neurology, Institut d'Investigacions Biomediques Sant Pau-Hospital Santa Creu i Sant Pau, Universitat Autonoma de Barcelona, 08025 Barcelona, Spain
| | - Alberto Lleo
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Department of Neurology, Institut d'Investigacions Biomediques Sant Pau-Hospital Santa Creu i Sant Pau, Universitat Autonoma de Barcelona, 08025 Barcelona, Spain
| | - Pascual Sanchez-Juan
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Service of Neurology, IDIVAL, University Hospital Marques de Valdecilla, University of Cantabria, 39008 Santander, Spain
| | - Juan Fortea
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain
- Department of Neurology, Institut d'Investigacions Biomediques Sant Pau-Hospital Santa Creu i Sant Pau, Universitat Autonoma de Barcelona, 08025 Barcelona, Spain
| |
Collapse
|
10
|
Roy J, Tsui KC, Ng J, Fung ML, Lim LW. Regulation of Melatonin and Neurotransmission in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22136841. [PMID: 34202125 PMCID: PMC8268832 DOI: 10.3390/ijms22136841] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/20/2022] Open
Abstract
Alzheimer’s disease is a neurodegenerative disorder associated with age, and is characterized by pathological markers such as amyloid-beta plaques and neurofibrillary tangles. Symptoms of AD include cognitive impairments, anxiety and depression. It has also been shown that individuals with AD have impaired neurotransmission, which may result from the accumulation of amyloid plaques and neurofibrillary tangles. Preclinical studies showed that melatonin, a monoaminergic neurotransmitter released from the pineal gland, is able to ameliorate AD pathologies and restore cognitive impairments. Theoretically, inhibition of the pathological progression of AD by melatonin treatment should also restore the impaired neurotransmission. This review aims to explore the impact of AD on neurotransmission, and whether and how melatonin can enhance neurotransmission via improving AD pathology.
Collapse
|
11
|
Azmi H. Neuromodulation for Cognitive Disorders: In Search of Lazarus? Neurol India 2021; 68:S288-S296. [PMID: 33318364 DOI: 10.4103/0028-3886.302469] [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: 11/04/2022]
Abstract
Alzheimer's disease (AD) and other forms of dementia can have a large impact on patients, their families, and for the society as a whole. Current medical treatments have not shown enough potential in treating or altering the course of the disease. Deep brain stimulation (DBS) has shown great neuromodulatory potential in Parkinson's disease, and there is a growing body of evidence for justifying its use in cognitive disorders. At the same time there is mounting interest at less invasive and alternative modes of neuromodulation for the treatment of AD. This manuscript is a brief review of the infrastructure of memory, the current understanding of the pathophysiology of AD, and the body of preclinical and clinical evidence for noninvasive and invasive neuromodulation modalities for the treatment of cognitive disorders and AD in particular.
Collapse
Affiliation(s)
- Hooman Azmi
- Department of Neurosurgery, Hackensack University Medical Center, Hackensack Meridian Health, Hackensack; New Jersey Brain and Spine Center, Oradell, New Jersey, USA
| |
Collapse
|
12
|
Ovsepian SV, O'Leary VB, Hoschl C, Zaborszky L. Integrated phylogeny of the human brain and pathobiology of Alzheimer's disease: A unifying hypothesis. Neurosci Lett 2021; 755:135895. [PMID: 33862141 DOI: 10.1016/j.neulet.2021.135895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 04/05/2021] [Accepted: 04/09/2021] [Indexed: 02/08/2023]
Abstract
The disproportionate evolutionary expansion of the human cerebral cortex with reinforcement of cholinergic innervations warranted a major rise in the functional and metabolic load of the conserved basal forebrain (BF) cholinergic system. Given that acetylcholine (ACh) regulates properties of the microtubule-associated protein (MAP) tau and promotes non-amyloidogenic processing of amyloid precursor protein (APP), growing neocortex predicts higher demands for ACh, while the emerging role of BF cholinergic projections in Aβ clearance infers greater exposure of source neurons and their innervation fields to amyloid pathology. The higher exposure of evolutionary most recent cortical areas to the amyloid pathology of Alzheimer's disease (AD) with synaptic impairments and atrophy, therefore, might involve attenuated homeostatic effects of BF cholinergic projections, in addition to fall-outs of inherent processes of expanding association areas. This unifying model, thus, views amyloid pathology and loss of cholinergic cells as a quid pro quo of the allometric evolution of the human brain, which in combination with increase in life expectancy overwhelm the fine homeostatic balance and trigger the disease process.
Collapse
Affiliation(s)
- Saak V Ovsepian
- Department of Experimental Neurobiology, National Institute of Mental Health, Topolová 748, 250 67, Klecany, Czech Republic.
| | - Valerie B O'Leary
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Ruská 87, 100 00, Praha 10, Czech Republic
| | - Cyril Hoschl
- Department of Experimental Neurobiology, National Institute of Mental Health, Topolová 748, 250 67, Klecany, Czech Republic; Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Ruská 87, 100 00, Prague 10, Czech Republic
| | - Laszlo Zaborszky
- Center for Molecular and Behavioral Neuroscience, Rutgers, the State University of New Jersey, Newark, NJ, USA
| |
Collapse
|
13
|
Shekari A, Fahnestock M. Cholinergic neurodegeneration in Alzheimer disease mouse models. HANDBOOK OF CLINICAL NEUROLOGY 2021; 182:191-209. [PMID: 34266592 DOI: 10.1016/b978-0-12-819973-2.00013-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Cholinergic signaling is critical for cognitive function. The basal forebrain is the major cholinergic output of the central nervous system. Degeneration of basal forebrain cholinergic neurons is a hallmark of Alzheimer's disease (AD). Mouse models are invaluable tools in disease research and have been used to study AD for over 25 years. However, animal models of AD vary greatly with respect to the degree of cholinergic degeneration observed. The following review will outline the most influential animal models of AD with an emphasis on the basal forebrain cholinergic system.
Collapse
Affiliation(s)
- Arman Shekari
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Margaret Fahnestock
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
14
|
Therapeutic potential of mangiferin in the treatment of various neuropsychiatric and neurodegenerative disorders. Neurochem Int 2020; 143:104939. [PMID: 33346032 DOI: 10.1016/j.neuint.2020.104939] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 11/02/2020] [Accepted: 12/12/2020] [Indexed: 12/19/2022]
Abstract
Xanthones are important chemical class of bioactive products that confers therapeutic benefits. Of several xanthones, mangiferin is known to be distributed widely across several fruits, vegetables and medicinal plants. Mangiferin has been shown to exert neuroprotective effects in both in-vitro and in-vivo models. Mangiferin attenuates cerebral infarction, cerebral edema, lipid peroxidation (MDA), neuronal damage, etc. Mangiferin further potentiate levels of endogenous antioxidants to confer protection against the oxidative stress inside the neurons. Mangiferin is involved in the regulation of various signaling pathways that influences the production and levels of proinflammatory cytokines in brain. Mangiferin cosunteracted the neurotoxic effect of amyloid-beta, MPTP, rotenone, 6-OHDA etc and confer protection to neurons. These evidence suggested that the mangiferin may be a potential therapeutic strategy for the treatment of various neurological disorders. The present review demonstrated the pharmacodynamics-pharmacokinetics of mangiferin and neurotherapeutic potential in several neurological disorders with underlying mechanisms.
Collapse
|
15
|
Neuron Loss in Alzheimer's Disease: Translation in Transgenic Mouse Models. Int J Mol Sci 2020; 21:ijms21218144. [PMID: 33143374 PMCID: PMC7663280 DOI: 10.3390/ijms21218144] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023] Open
Abstract
Transgenic mouse models represent an essential tool for the exploration of Alzheimer’s disease (AD) pathological mechanisms and the development of novel treatments, which at present provide only symptomatic and transient effects. While a variety of mouse models successfully reflects the main neuropathological hallmarks of AD, such as extracellular amyloid-β (Aβ) deposits, intracellular accumulation of Tau protein, the development of micro- and astrogliosis, as well as behavioral deficits, substantial neuron loss, as a key feature of the disease, seems to be more difficult to achieve. In this review, we summarize information on classic and more recent transgenic mouse models for AD, focusing in particular on loss of pyramidal, inter-, and cholinergic neurons. Although the cause of neuron loss in AD is still a matter of scientific debate, it seems to be linked to intraneuronal Aβ accumulation in several transgenic mouse models, especially in pyramidal neurons.
Collapse
|
16
|
Wong KY, Roy J, Fung ML, Heng BC, Zhang C, Lim LW. Relationships between Mitochondrial Dysfunction and Neurotransmission Failure in Alzheimer's Disease. Aging Dis 2020; 11:1291-1316. [PMID: 33014538 PMCID: PMC7505271 DOI: 10.14336/ad.2019.1125] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022] Open
Abstract
Besides extracellular deposition of amyloid beta and formation of phosphorylated tau in the brains of patients with Alzheimer's disease (AD), the pathogenesis of AD is also thought to involve mitochondrial dysfunctions and altered neurotransmission systems. However, none of these components can describe the diverse cognitive, behavioural, and psychiatric symptoms of AD without the pathologies interacting with one another. The purpose of this review is to understand the relationships between mitochondrial and neurotransmission dysfunctions in terms of (1) how mitochondrial alterations affect cholinergic and monoaminergic systems via disruption of energy metabolism, oxidative stress, and apoptosis; and (2) how different neurotransmission systems drive mitochondrial dysfunction via increasing amyloid beta internalisation, oxidative stress, disruption of mitochondrial permeabilisation, and mitochondrial trafficking. All these interactions are separately discussed in terms of neurotransmission systems. The association of mitochondrial dysfunctions with alterations in dopamine, norepinephrine, and histamine is the prospective goal in this research field. By unfolding the complex interactions surrounding mitochondrial dysfunction in AD, we can better develop potential treatments to delay, prevent, or cure this devastating disease.
Collapse
Affiliation(s)
- Kan Yin Wong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Jaydeep Roy
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Man Lung Fung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Boon Chin Heng
- Peking University School of Stomatology, Beijing, China.
| | - Chengfei Zhang
- Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China.
| | - Lee Wei Lim
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
17
|
Sun JY. Anesthesia and Alzheimer's: A review. J Anaesthesiol Clin Pharmacol 2020; 36:297-302. [PMID: 33487895 PMCID: PMC7812964 DOI: 10.4103/joacp.joacp_118_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 09/21/2019] [Accepted: 10/28/2019] [Indexed: 11/28/2022] Open
Abstract
As early as 1955, it was Bedford who provided description of cognitive changes in elderly patients following anesthesia and surgery. Reports of individuals with catastrophic, non-stroke-related decline in cognitive functions following anesthesia and surgery lead to a perception in the lay population that anesthesia and surgery have the potential to greatly exaggerate the progression of dementia, particularly Alzheimer's disease (AD). There is a concern that anesthesia and surgery could cause irreversible impairment, leading to AD. This could also explain the accelerated decline in patients with mild cognitive impairment. We seek to explore the relevant literature to determine whether a correlation exists and then propose a possible pathophysiologic mechanism.
Collapse
Affiliation(s)
- Jeffrey Y Sun
- NYU Langone Health, Department of Anesthesiology, Perioperative Care and Pain Medicine, New York, New York, USA
| |
Collapse
|
18
|
Kosel F, Pelley JMS, Franklin TB. Behavioural and psychological symptoms of dementia in mouse models of Alzheimer's disease-related pathology. Neurosci Biobehav Rev 2020; 112:634-647. [PMID: 32070692 DOI: 10.1016/j.neubiorev.2020.02.012] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 02/08/2020] [Accepted: 02/11/2020] [Indexed: 12/12/2022]
Abstract
Transgenic mouse models have been used extensively to model the cognitive impairments arising from Alzheimer's disease (AD)-related pathology. However, less is known about the relationship between AD-related pathology and the behavioural and psychological symptoms of dementia (BPSD) commonly presented by patients. This review discusses the BPSD-like behaviours recapitulated by several mouse models of AD-related pathology, including the APP/PS1, Tg2576, 3xTg-AD, 5xFAD, and APP23 models. Current evidence suggests that social withdrawal and depressive-like behaviours increase with progressive neuropathology, and increased aggression and sleep-wake disturbances are present even at early stages; however, there is no clear evidence to support increased anxiety-like behaviours, agitation (hyperactivity), or general apathy. Overall, transgenic mouse models of AD-related pathology recapitulate some of the BPSD-like behaviours associated with AD, but these behaviours vary by model. This reflects the patient population, where AD patients typically exhibit one or more BPSD, but rarely all symptoms at once. As a result, we suggest that transgenic mouse models are an important tool to investigate the pathology underlying BPSD in human AD patients.
Collapse
Affiliation(s)
- Filip Kosel
- The Social Lab, Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Jessica M S Pelley
- The Social Lab, Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Tamara B Franklin
- The Social Lab, Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada.
| |
Collapse
|
19
|
Shekarian M, Komaki A, Shahidi S, Sarihi A, Salehi I, Raoufi S. The protective and therapeutic effects of vinpocetine, a PDE1 inhibitor, on oxidative stress and learning and memory impairment induced by an intracerebroventricular (ICV) injection of amyloid beta (aβ) peptide. Behav Brain Res 2020; 383:112512. [PMID: 31991177 DOI: 10.1016/j.bbr.2020.112512] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease leading to cognitive and memory impairment. This study aimed at investigating the therapeutic and preserving effects of vinpocetine on amyloid beta (Aβ)-induced rat model of AD. Sixty male adult Wistar rats were randomly divided into 6 groups (n = 10 per group) as follows: 1; control, 2; sham, 3; Aβ, 4; pre-treatment (vinpocetine + Aβ): oral gavage administration of vinpocetine at 4 mg/kg for 30 days followed by intracerebroventricular (ICV) injection of Aβ, 5; treatment (Aβ + vinpocetine): Aβ ICV injection followed by vinpocetine administration for 30 days, 6; pre-treatment + treatment (vinpocetine + Aβ + vinpocetine): vinpocetine administration for 30 days before and 30 days after AD induction. Following treatments, the animals' learning and memory were investigated using passive avoidance learning (PAL) task, Morris water maze (MWM), and novel object recognition (NOR) tests. The results demonstrated that Aβ significantly enhanced escape latency and the distance traveled in the MWM, decreased step-through latency, and increased time spent in the dark compartment in PAL. Vinpocetine ameliorated the Aβ-infused memory deficits in both MWM and PAL tests. Administration of vinpocetine in the Aβ rats increased the discrimination index of the NOR test. It also significantly diminished the nitric oxide and malondialdehyde levels and restored the reduced glutathione (GSH) levels. Vinpocetine can improve memory and learning impairment following Aβ infusion due to its different properties, including antioxidant effects, which indicates that vinpocetine administration can lead to the amelioration of cognitive dysfunction in AD.
Collapse
Affiliation(s)
- Meysam Shekarian
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Siamak Shahidi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abdolrahman Sarihi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Salehi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Safoura Raoufi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
20
|
Alzheimer's disease: Neurotransmitters of the sleep-wake cycle. Neurosci Biobehav Rev 2019; 105:72-80. [PMID: 31377219 DOI: 10.1016/j.neubiorev.2019.07.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 05/22/2019] [Accepted: 07/28/2019] [Indexed: 11/22/2022]
Abstract
With aging, our sleeping pattern alters. Elderly often wake unrested because their sleep time and sleep efficacy is reduced. In Alzheimer's disease (AD) patients, these alterations are even more pronounced and may further aggravate cognitive decline. Therefore, sleep disturbances greatly impact self-care ability, caregiver exhaustion and institutionalization rate. Reestablishing an effective sleep-wake cycle in these patients still remains an unresolved challenge, partly because sleep physiology is quite complex and multiple neurotransmitter systems contribute to a single process. Gaining a better understanding of sleep physiology will be crucial for further research. Conjointly, animal models, along with a multidisciplinary approach, will be of great value to establish a common ground between AD and sleep disturbances and work towards a potential therapeutic application.
Collapse
|
21
|
Reale M, D'Angelo C, Costantini E, Di Nicola M, Yarla NS, Kamal MA, Salvador N, Perry G. Expression Profiling of Cytokine, Cholinergic Markers, and Amyloid-β Deposition in the APPSWE/PS1dE9 Mouse Model of Alzheimer's Disease Pathology. J Alzheimers Dis 2019; 62:467-476. [PMID: 29439355 PMCID: PMC5817902 DOI: 10.3233/jad-170999] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Alzheimer’s disease (AD), a neurodegenerative disease, is associated with dysfunction of the olfactory and the entorhinal cortex of the brain that control memory and cognitive functions and other daily activities. Pro-inflammatory cytokines, amyloid-β (Aβ), and the cholinergic system play vital roles in the pathophysiology of AD. However, the role of changes in cholinergic system components, Aβ accumulation, and cytokines in both the olfactory and entorhinal cortex is not known clearly. Objective: The present study is aimed to evaluate the changes of cholinergic system components, Aβ accumulation, and cytokines in both the olfactory bulb (OB) and entorhinal cortex (EC) of young and aged APPSWE/PS1dE9 transgenic (Tg) mice. Methods: We have explored the changes of cholinergic system components, Aβ accumulation, and expression profiling of cytokines in the OB and EC of aged APPswe transgenic mice and age-matched wild type mice using quantitative Real-Time PCR assays and immunohistochemistry techniques. Results: In aged Tg mice, a significant increase of expression of interleukin (IL)-1β, tumor necrosis factor (TNF)-α, and chemokine MCP1 (p < 0.001, p < 0.001, and p = 0.001, respectively) and a significant reduction of nAChRα4 (p = 0.048) and AChE (p = 0.023) was observed when compared with age-matched wild type mice. Higher levels of AChE and BuChE are expressed in OB and EC of the APPSWE/PS1dE9 of Tg mice. Aβ accumulation was observed in OB and EC of the APPSWE/PS1dE9 of Tg mice. Conclusion: The study demonstrates the expression profiling of pro-inflammatory cytokines and cholinergic markers as well as Aβ accumulation in OB and EC of the APPSWE/PS1dE9 Tg mice. Moreover, the study also demonstrated that the APPSWE/PS1dE9 Tg mice can be useful as a mouse model to understand the role of pro-inflammatory cytokines and cholinergic markers in pathophysiology of AD.
Collapse
Affiliation(s)
- Marcella Reale
- Department of Medical, Oral and Biotechnological Sciences, University "G. D'Annunzio", Chieti, Italy
| | - Chiara D'Angelo
- Department of Medical, Oral and Biotechnological Sciences, University "G. D'Annunzio", Chieti, Italy
| | - Erica Costantini
- Department of Medical, Oral and Biotechnological Sciences, University "G. D'Annunzio", Chieti, Italy
| | - Marta Di Nicola
- Department of Medical, Oral and Biotechnological Sciences, University "G. D'Annunzio", Chieti, Italy
| | - Nagnedra Sastry Yarla
- Department of Physiology, Divisions of Chemistry and Biochemistry, Pharmacology and Neuroscience, City University of New York Medical School, New York, NY, USA
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Enzymoics, Hebersham, NSW, Australia.,Novel Global Community Educational Foundation, Australia
| | - Nieves Salvador
- Department of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal-CSIC, Madrid, Spain
| | - George Perry
- Department of Biology, College of Sciences, The University of Texas at San Antonio, San Antonio, TX, USA
| |
Collapse
|
22
|
Xiong L, Duan L, Xu W, Wang Z. Nerve growth factor metabolic dysfunction contributes to sevoflurane-induced cholinergic degeneration and cognitive impairments. Brain Res 2018; 1707:107-116. [PMID: 30481505 DOI: 10.1016/j.brainres.2018.11.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 11/21/2018] [Accepted: 11/24/2018] [Indexed: 12/26/2022]
Abstract
General anesthesia with sevoflurane is associated with an increased incidence of postoperative cognitive dysfunction. Previous studies have shown that sevoflurane anesthesia can affect the integrity and function of basal forebrain cholinergic neurons (BFCNs) which are essential for learning and memory. However, the underlying mechanisms remain largely unknown. Here, we demonstrated that exposure to 2.5% sevoflurane induced significant loss of BFCNs and caused impairments of the spatial and the fear memory. Further, sevoflurane exposure significantly reduced the level of nerve growth factor (NGF), an important factor for the survival and phenotype maintenance of BFCNs, by disrupting its synthesis pathways in the brain. More importantly, NGF administration not only prevented the loss of BFCNs but also ameliorated the cognitive impairments in sevoflurane-treated mice. Our findings indicate that NGF metabolic dysfunction contributes to sevoflurane-associated BFCNs degeneration and subsequent cognitive deficits.
Collapse
Affiliation(s)
- Lu Xiong
- Department of Anesthesiology, Tinglin Hospital of Jinshan Disctrict, Shanghai 201505, China
| | - Lijie Duan
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Wenqing Xu
- Department of Anesthesiology, Tinglin Hospital of Jinshan Disctrict, Shanghai 201505, China
| | - Zigao Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
23
|
Grothe MJ, Kilimann I, Grinberg L, Heinsen H, Teipel S. In Vivo Volumetry of the Cholinergic Basal Forebrain. NEUROMETHODS 2018. [DOI: 10.1007/978-1-4939-7674-4_15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
24
|
Kaur G, Pawlik M, Gandy SE, Ehrlich ME, Smiley JF, Levy E. Lysosomal dysfunction in the brain of a mouse model with intraneuronal accumulation of carboxyl terminal fragments of the amyloid precursor protein. Mol Psychiatry 2017; 22:981-989. [PMID: 27777419 PMCID: PMC5405008 DOI: 10.1038/mp.2016.189] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/05/2016] [Accepted: 08/25/2016] [Indexed: 11/12/2022]
Abstract
Recent data suggest that intraneuronal accumulation of metabolites of the amyloid-β-precursor protein (APP) is neurotoxic. We observed that transgenic mice overexpressing in neurons a human APP gene harboring the APPE693Q (Dutch) mutation have intraneuronal lysosomal accumulation of APP carboxylterminal fragments (APP-CTFs) and oligomeric amyloid β (oAβ) but no histological evidence of amyloid deposition. Morphometric quantification using the lysosomal marker protein 2 (LAMP-2) immunolabeling showed higher neuronal lysosomal counts in brain of 12-months-old APPE693Q as compared with age-matched non-transgenic littermates, and western blots showed increased lysosomal proteins including LAMP-2, cathepsin D and LC3. At 24 months of age, these mice also exhibited an accumulation of α-synuclein in the brain, along with increased conversion of LC3-I to LC3-II, an autophagosomal/autolysosomal marker. In addition to lysosomal changes at 12 months of age, these mice developed cholinergic neuronal loss in the basal forebrain, GABAergic neuronal loss in the cortex, hippocampus and basal forebrain and gliosis and microgliosis in the hippocampus. These findings suggest a role for the intraneuronal accumulation of oAβ and APP-CTFs and resultant lysosomal pathology at early stages of Alzheimer's disease-related pathology.
Collapse
Affiliation(s)
| | | | - Sam E. Gandy
- Departments of Neurology and Psychiatry, and Alzheimer’s Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, and James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
| | - Michelle E. Ehrlich
- Departments of Neurology and Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John F. Smiley
- Nathan S. Kline Institute, Orangeburg, NY, USA,Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA
| | - Efrat Levy
- Nathan S. Kline Institute, Orangeburg, NY, USA,Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA,Department of Biochemistry and Molecular Pharmacology, NYU Langone Medical Center, New York, NY, USA
| |
Collapse
|
25
|
Canu N, Amadoro G, Triaca V, Latina V, Sposato V, Corsetti V, Severini C, Ciotti MT, Calissano P. The Intersection of NGF/TrkA Signaling and Amyloid Precursor Protein Processing in Alzheimer's Disease Neuropathology. Int J Mol Sci 2017. [PMID: 28632177 PMCID: PMC5486140 DOI: 10.3390/ijms18061319] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Dysfunction of nerve growth factor (NGF) and its high-affinity Tropomyosin receptor kinase A (TrkA) receptor has been suggested to contribute to the selective degeneration of basal forebrain cholinergic neurons (BFCN) associated with the progressive cognitive decline in Alzheimer's disease (AD). The aim of this review is to describe our progress in elucidating the molecular mechanisms underlying the dynamic interplay between NGF/TrkA signaling and amyloid precursor protein (APP) metabolism within the context of AD neuropathology. This is mainly based on the finding that TrkA receptor binding to APP depends on a minimal stretch of ~20 amino acids located in the juxtamembrane/extracellular domain of APP that carries the α- and β-secretase cleavage sites. Here, we provide evidence that: (i) NGF could be one of the “routing” proteins responsible for modulating the metabolism of APP from amyloidogenic towards non-amyloidogenic processing via binding to the TrkA receptor; (ii) the loss of NGF/TrkA signaling could be linked to sporadic AD contributing to the classical hallmarks of the neuropathology, such as synaptic loss, β-amyloid peptide (Aβ) deposition and tau abnormalities. These findings will hopefully help to design therapeutic strategies for AD treatment aimed at preserving cholinergic function and anti-amyloidogenic activity of the physiological NGF/TrkA pathway in the septo-hippocampal system.
Collapse
Affiliation(s)
- Nadia Canu
- Department of System Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00137 Rome, Italy.
- Institute of Cellular Biology and Neurobiology, National Council of Research Rome, Via del Fosso del Fiorano 64, 00143 Rome, Italy.
| | - Giuseppina Amadoro
- Institute of Translational Pharmacology, National Research Council (CNR) Rome, Via Fosso del Cavaliere 100, 00133 Rome, Italy.
| | - Viviana Triaca
- Institute of Cellular Biology and Neurobiology, National Council of Research Rome, Via del Fosso del Fiorano 64, 00143 Rome, Italy.
| | - Valentina Latina
- Institute of Translational Pharmacology, National Research Council (CNR) Rome, Via Fosso del Cavaliere 100, 00133 Rome, Italy.
| | - Valentina Sposato
- European Brain Research Institute Rome, Via del Fosso del Fiorano 64, 00143 Rome, Italy.
| | - Veronica Corsetti
- European Brain Research Institute Rome, Via del Fosso del Fiorano 64, 00143 Rome, Italy.
| | - Cinzia Severini
- Institute of Cellular Biology and Neurobiology, National Council of Research Rome, Via del Fosso del Fiorano 64, 00143 Rome, Italy.
| | - Maria Teresa Ciotti
- Institute of Cellular Biology and Neurobiology, National Council of Research Rome, Via del Fosso del Fiorano 64, 00143 Rome, Italy.
| | - Pietro Calissano
- European Brain Research Institute Rome, Via del Fosso del Fiorano 64, 00143 Rome, Italy.
| |
Collapse
|
26
|
Teipel SJ, Cavedo E, Weschke S, Grothe MJ, Rojkova K, Fontaine G, Dauphinot L, Gonzalez-Escamilla G, Potier MC, Bertin H, Habert MO, Dubois B, Hampel H. Cortical amyloid accumulation is associated with alterations of structural integrity in older people with subjective memory complaints. Neurobiol Aging 2017. [PMID: 28646687 DOI: 10.1016/j.neurobiolaging.2017.05.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We determined the effect of cortical amyloid load using 18F-florbetapir PET on cognitive performance and gray matter structural integrity derived from MRI in 318 cognitively normally performing older people with subjective memory impairment from the INSIGHT-preAD cohort using multivariate partial least squares regression. Amyloid uptake was associated with reduced gray matter structural integrity in hippocampus, entorhinal and cingulate cortex, middle temporal gyrus, prefrontal cortex, and lentiform nucleus (p < 0.01, permutation test). Higher amyloid load was associated with poorer global cognitive performance, delayed recall and attention (p < 0.05), independently of its effects on gray matter connectivity. These findings agree with the assumption of a two-stage effect of amyloid on cognition, (1) an early direct effect in the preclinical stages of Alzheimer's disease and (2) a delayed effect mediated by downstream effects of amyloid accumulation, such as gray matter connectivity decline.
Collapse
Affiliation(s)
- Stefan J Teipel
- German Center for Neurodegenerative Diseases (DZNE) - Rostock/Greifswald, Rostock, Germany; Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany.
| | - Enrica Cavedo
- AXA Research Fund & UPMC Chair, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du cerveau et de la moelle (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l'hôpital, Paris, France; IRCCS Istituto Centro San Giovanni di Dio-Fatebenefratelli, Brescia, Italy
| | - Sarah Weschke
- German Center for Neurodegenerative Diseases (DZNE) - Rostock/Greifswald, Rostock, Germany; Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| | - Michel J Grothe
- German Center for Neurodegenerative Diseases (DZNE) - Rostock/Greifswald, Rostock, Germany; Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| | - Katrine Rojkova
- AXA Research Fund & UPMC Chair, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du cerveau et de la moelle (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l'hôpital, Paris, France
| | - Gaëlle Fontaine
- ICM Institut du Cerveau et de la Moelle épinière, CNRS UMR7225, INSERM U1127, UPMC, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Luce Dauphinot
- ICM Institut du Cerveau et de la Moelle épinière, CNRS UMR7225, INSERM U1127, UPMC, Hôpital de la Pitié-Salpêtrière, Paris, France
| | | | - Marie-Claude Potier
- ICM Institut du Cerveau et de la Moelle épinière, CNRS UMR7225, INSERM U1127, UPMC, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Hugo Bertin
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, Paris, France; Centre pour l'Acquisition et le Traitement des Images, Paris, France
| | - Marie-Odile Habert
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, Paris, France; Centre pour l'Acquisition et le Traitement des Images, Paris, France; AP-HP, Hôpital Pitié-Salpêtrière, Département de Médecine Nucléaire, Paris, France
| | - Bruno Dubois
- Sorbonne Universités, UPMC Univ Paris 06, Inserm, CNRS, Institut du cerveau et de la moelle (ICM) - Hôpital Pitié-Salpêtrière, Boulevard de l'hôpital, Paris, France
| | - Harald Hampel
- AXA Research Fund & UPMC Chair, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du cerveau et de la moelle (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l'hôpital, Paris, France
| | | |
Collapse
|
27
|
Latina V, Caioli S, Zona C, Ciotti MT, Amadoro G, Calissano P. Impaired NGF/TrkA Signaling Causes Early AD-Linked Presynaptic Dysfunction in Cholinergic Primary Neurons. Front Cell Neurosci 2017; 11:68. [PMID: 28360840 PMCID: PMC5350152 DOI: 10.3389/fncel.2017.00068] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 02/24/2017] [Indexed: 12/31/2022] Open
Abstract
Alterations in NGF/TrkA signaling have been suggested to underlie the selective degeneration of the cholinergic basal forebrain neurons occurring in vivo in AD (Counts and Mufson, 2005; Mufson et al., 2008; Niewiadomska et al., 2011) and significant reduction of cognitive decline along with an improvement of cholinergic hypofunction have been found in phase I clinical trial in humans affected from mild AD following therapeutic NGF gene therapy (Tuszynski et al., 2005, 2015). Here, we show that the chronic (10–12 D.I.V.) in vitro treatment with NGF (100 ng/ml) under conditions of low supplementation (0.2%) with the culturing serum-substitute B27 selectively enriches the basal forebrain cholinergic neurons (+36.36%) at the expense of other non-cholinergic, mainly GABAergic (−38.45%) and glutamatergic (−56.25%), populations. By taking advantage of this newly-developed septo-hippocampal neuronal cultures, our biochemical and electrophysiological investigations demonstrate that the early failure in excitatory neurotransmission following NGF withdrawal is paralleled by concomitant and progressive loss in selected presynaptic and vesicles trafficking proteins including synapsin I, SNAP-25 and α-synuclein. This rapid presynaptic dysfunction: (i) precedes the commitment to cell death and is reversible in a time-dependent manner, being suppressed by de novo external administration of NGF within 6 hr from its initial withdrawal; (ii) is specific because it is not accompanied by contextual changes in expression levels of non-synaptic proteins from other subcellular compartments; (ii) is not secondary to axonal degeneration because it is insensible to pharmacological treatment with known microtubule-stabilizing drug such paclitaxel; (iv) involves TrkA-dependent mechanisms because the effects of NGF reapplication are blocked by acute exposure to specific and cell-permeable inhibitor of its high-affinity receptor. Taken together, this study may have important clinical implications in the field of AD neurodegeneration because it: (i) provides new insights on the earliest molecular mechanisms underlying the loss of synaptic/trafficking proteins and, then, of synapes integrity which occurs in vulnerable basal forebrain population at preclinical stages of neuropathology; (ii) offers prime presynaptic-based molecular target to extend the therapeutic time-window of NGF action in the strategy of improving its neuroprotective in vivo intervention in affected patients.
Collapse
Affiliation(s)
- Valentina Latina
- Institute of Translational Pharmacology, National Research Council (CNR) Rome, Italy
| | | | - Cristina Zona
- IRCCS Santa Lucia FoundationRome, Italy; Department of Systems Medicine, University of Rome Tor VergataRome, Italy
| | - Maria T Ciotti
- NGF and Molecular Mechanisms of Neurodegenerative Diseases, European Brain Research Institute (EBRI) Rome, Italy
| | - Giuseppina Amadoro
- Institute of Translational Pharmacology, National Research Council (CNR)Rome, Italy; NGF and Molecular Mechanisms of Neurodegenerative Diseases, European Brain Research Institute (EBRI)Rome, Italy
| | - Pietro Calissano
- NGF and Molecular Mechanisms of Neurodegenerative Diseases, European Brain Research Institute (EBRI) Rome, Italy
| |
Collapse
|
28
|
Foidl BM, Do-Dinh P, Hutter-Schmid B, Bliem HR, Humpel C. Cholinergic neurodegeneration in an Alzheimer mouse model overexpressing amyloid-precursor protein with the Swedish-Dutch-Iowa mutations. Neurobiol Learn Mem 2016; 136:86-96. [PMID: 27670619 PMCID: PMC6020032 DOI: 10.1016/j.nlm.2016.09.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 08/26/2016] [Accepted: 09/19/2016] [Indexed: 10/21/2022]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder that is mainly characterized by beta-amyloid (Aβ) plaque deposition, Tau pathology and dysfunction of the cholinergic system causing memory impairment. The aim of the present study was to examine (1) anxiety and cognition, (2) Aβ plaque deposition and (3) degeneration of cholinergic neurons in the nucleus basalis of Meynert (nbM) and cortical cholinergic innervation in an Alzheimer mouse model (APP_SweDI; overexpressing amyloid precursor protein (APP) with the Swedish K670N/M671L, Dutch E693Q, and Iowa D694N mutations). Our results show that 12-month-old APP_SweDI mice were more anxious and had more memory impairment. A large number of Aβ plaques were already visible at the age of 6 months and increased with age. A significant decrease in cholinergic neurons was seen in the transgenic mouse model in comparison to the wild-type mice, identified by immunohistochemistry against choline acetyltransferase (ChAT) and p75 neurotrophin receptor as well as by in situ hybridization. Moreover, a significant decrease in cortical cholinergic fiber density was found in the transgenic mice as compared to the wild-type. In the cerebral cortex of APP_SweDI mice, swollen cholinergic varicosities were seen in the vicinity of Aβ plaques. In conclusion, the present study shows that in an AD mouse model (APP_SweDI mice) a high Aβ plaque load in the cortex causes damage to cholinergic axons in the cortex, followed by subsequent retrograde-induced cell death of cholinergic neurons and some forms of compensatory processes. This degeneration was accompanied by enhanced anxiety and impaired cognition.
Collapse
Affiliation(s)
- Bettina Maria Foidl
- Laboratory of Psychiatry and Exp. Alzheimer's Research, Department of Psychiatry and Psychotherapy, Medical University of Innsbruck, Austria
| | - Patricia Do-Dinh
- Laboratory of Psychiatry and Exp. Alzheimer's Research, Department of Psychiatry and Psychotherapy, Medical University of Innsbruck, Austria
| | - Bianca Hutter-Schmid
- Laboratory of Psychiatry and Exp. Alzheimer's Research, Department of Psychiatry and Psychotherapy, Medical University of Innsbruck, Austria
| | - Harald R Bliem
- Laboratory of Psychiatry and Exp. Alzheimer's Research, Department of Psychiatry and Psychotherapy, Medical University of Innsbruck, Austria; Department of Psychology, University of Innsbruck, Austria
| | - Christian Humpel
- Laboratory of Psychiatry and Exp. Alzheimer's Research, Department of Psychiatry and Psychotherapy, Medical University of Innsbruck, Austria.
| |
Collapse
|
29
|
Janssen L, Dubbelaar ML, Holtman IR, de Boer-Bergsma J, Eggen BJL, Boddeke HWGM, De Deyn PP, Van Dam D. Aging, microglia and cytoskeletal regulation are key factors in the pathological evolution of the APP23 mouse model for Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2016; 1863:395-405. [PMID: 27838490 DOI: 10.1016/j.bbadis.2016.11.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 10/21/2016] [Accepted: 11/08/2016] [Indexed: 02/01/2023]
Abstract
Aging is the key risk factor for Alzheimer's disease (AD). In addition, the amyloid-beta (Aβ) peptide is considered a critical neurotoxic agent in AD pathology. However, the connection between these factors is unclear. We aimed to provide an extensive characterization of the gene expression profiles of the amyloidosis APP23 model for AD and control mice and to evaluate the effect of aging on these profiles. We also correlated our findings to changes in soluble Aβ-levels and other pathological and symptomatic features of the model. We observed a clear biphasic expression profile. The first phase displayed a maturation profile, which resembled features found in young carriers of familial AD mutations. The second phase reflected aging processes and showed similarities to the progression of human AD pathology. During this phase, the model displayed a clear upregulation of microglial activation and lysosomal pathways and downregulation of neuron differentiation and axon guidance pathways. Interestingly, the changes in expression were all correlated to aging in general, but appeared more extensive/accelerated in APP23 mice.
Collapse
Affiliation(s)
- Leen Janssen
- Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Marissa L Dubbelaar
- Department of Neuroscience, Medical Physiology Section, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Inge R Holtman
- Department of Neuroscience, Medical Physiology Section, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Jelkje de Boer-Bergsma
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Bart J L Eggen
- Department of Neuroscience, Medical Physiology Section, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Hendrikus W G M Boddeke
- Department of Neuroscience, Medical Physiology Section, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Peter P De Deyn
- Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium; University of Groningen, University Medical Center Groningen (UMCG), Department of Neurology and Alzheimer Research Center, Groningen, The Netherlands; Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Debby Van Dam
- Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium.
| |
Collapse
|
30
|
Maternal separation exacerbates Alzheimer's disease-like behavioral and pathological changes in adult APPswe/PS1dE9 mice. Behav Brain Res 2016; 318:18-23. [PMID: 27771383 DOI: 10.1016/j.bbr.2016.10.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 10/17/2016] [Accepted: 10/19/2016] [Indexed: 11/20/2022]
Abstract
Alzheimer's disease (AD), the most common neurodegenerative disorder that gradually destroys memory and cognitive abilities in the elderly, makes a huge emotional and economic burden on the patients and their families. The presence of senile plaques and the loss of cholinergic neurons in the brain are two neuropathological hallmarks of AD. Maternal separation (MS) is an animal paradigm designed to make early life stress. Studies on wild type rodents showed that MS could induce AD-like cognitive deficit and pathological changes. However, the effects of MS on AD susceptible population or AD animal models are still unclear. In the present study, male APPswe/PS1dE9 transgenic mice were separated from dam and pups 3h per day from postnatal day 2 to day 21. After weaning, all animals were housed under normal conditions (4 mice per cage). At 9-month age, MWM tests were performed to evaluate the learning and memory abilities. Then the pathological changes in the brain were measured by histology staining. The results showed MS mice had more severe deficit of learning and memory. Compared to the control, there were more senile plaques in cortex and hippocampus, fewer cholinergic neurons in nucleus basalis of Meynert in MS mice. These results indicate that MS exacerbates Alzheimer's disease-like behavioral and pathological changes in APPswe/PS1dE9 mice.
Collapse
|
31
|
Deibel S, Weishaupt N, Regis A, Hong N, Keeley R, Balog R, Bye C, Himmler S, Whitehead S, McDonald R. Subtle learning and memory impairment in an idiopathic rat model of Alzheimer's disease utilizing cholinergic depletions and β-amyloid. Brain Res 2016; 1646:12-24. [DOI: 10.1016/j.brainres.2016.05.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 02/02/2023]
|
32
|
Brueggen K, Dyrba M, Barkhof F, Hausner L, Filippi M, Nestor PJ, Hauenstein K, Klöppel S, Grothe MJ, Kasper E, Teipel SJ. Basal Forebrain and Hippocampus as Predictors of Conversion to Alzheimer's Disease in Patients with Mild Cognitive Impairment - A Multicenter DTI and Volumetry Study. J Alzheimers Dis 2016; 48:197-204. [PMID: 26401940 DOI: 10.3233/jad-150063] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Hippocampal grey matter (GM) atrophy predicts conversion from mild cognitive impairment (MCI) to Alzheimer's disease (AD). Pilot data suggests that mean diffusivity (MD) in the hippocampus, as measured with diffusion tensor imaging (DTI), may be a more accurate predictor of conversion than hippocampus volume. In addition, previous studies suggest that volume of the cholinergic basal forebrain may reach a diagnostic accuracy superior to hippocampal volume in MCI. OBJECTIVE The present study investigated whether increased MD and decreased volume of the hippocampus, the basal forebrain and other AD-typical regions predicted time to conversion from MCI to AD dementia. METHODS 79 MCI patients with DTI and T1-weighted magnetic resonance imaging (MRI) were retrospectively included from the European DTI Study in Dementia (EDSD) dataset. Of these participants, 35 converted to AD dementia after 6-46 months (mean: 21 months). We used Cox regression to estimate the relative conversion risk predicted by MD values and GM volumes, controlling for age, gender, education and center. RESULTS Decreased GM volume in all investigated regions predicted an increased risk for conversion. Additionally, increased MD in the right basal forebrain predicted increased conversion risk. Reduced volume of the right hippocampus was the only significant predictor in a stepwise model combining all predictor variables. CONCLUSION Volume reduction of the hippocampus, the basal forebrain and other AD-related regions was predictive of increased risk for conversion from MCI to AD. In this study, volume was superior to MD in predicting conversion.
Collapse
Affiliation(s)
| | - Martin Dyrba
- DZNE, German Center for Neurodegenerative Diseases, Rostock, Germany.,MMIS group, University of Rostock, Rostock, Germany
| | - Frederik Barkhof
- Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, Netherlands
| | - Lucrezia Hausner
- Department of Geriatric Psychiatry, Zentralinstitut für Seelische Gesundheit Mannheim, University of Heidelberg, Mannheim, Germany
| | - Massimo Filippi
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, Scientific Institute and University Vita-Salute San Raffaele, Milano, Italy
| | - Peter J Nestor
- DZNE, German Center for Neurodegenerative Diseases, Magdeburg, Germany
| | | | - Stefan Klöppel
- Department of Psychiatry and Psychotherapy, Freiburg Brain Imaging, University Clinic Freiburg, Freiburg, Germany
| | - Michel J Grothe
- DZNE, German Center for Neurodegenerative Diseases, Rostock, Germany
| | - Elisabeth Kasper
- Department of Psychosomatic Medicine, University Medicine Rostock, Rostock, Germany
| | - Stefan J Teipel
- DZNE, German Center for Neurodegenerative Diseases, Rostock, Germany.,Department of Psychosomatic Medicine, University Medicine Rostock, Rostock, Germany
| |
Collapse
|
33
|
Conti E, Tremolizzo L, Santarone ME, Tironi M, Radice I, Zoia CP, Aliprandi A, Salmaggi A, Dominici R, Casati M, Appollonio I, Ferrarese C. Donepezil modulates the endogenous immune response: implications for Alzheimer's disease. Hum Psychopharmacol 2016; 31:296-303. [PMID: 27297668 DOI: 10.1002/hup.2538] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 03/18/2016] [Accepted: 04/14/2016] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Donepezil (DNPZ) is a drug commonly used for Alzheimer's disease (AD) that may favour a T helper 2 phenotype leading to increased naturally occurring auto-antibodies (NAb) against beta-amyloid (Aβ). We hypothesized the involvement of the cholinergic receptors [α7-nicotnic acetylcholine receptor (α7nAChR)] expressed on peripheral blood mononuclear cells (PBMC). METHODS Fifty patients with mild-to-moderate AD, DNPZ treated (DNPZ+, n = 25) or not (DNPZ-, n = 25), and 25 matched controls were enrolled and PBMC extracted for both in vitro cultures, and real-time polymerase chain reaction and chromatin immunoprecipitation assay. Plasma samples were also obtained for Aβ and NAb determination. RESULTS Donepezil increased in vitro the expression of the transcription factor GATA binding protein 3 (GATA3) through α7nAChR, because prevented by the specific antagonist methyllycaconitine. Ex vivo PBMC α7nAChR mRNA expression was increased in both AD groups, while GATA3 expression was not. A significant increase in the GATA3/interleukin 5 promoter association was found in DNPZ+ patients. Finally, DNPZ+ patients showed both significantly higher plasma levels of anti-Aβ NAb with respect to DNPZ- patients and Aβ 1-42 with respect to normal controls. CONCLUSIONS Donepezil might modulate a T helper 2 bias via α7nAChR leading to increased expression of NAb. Further studies on the role of the modulation of the immune response against Aβ may pave the way to innovative therapeutic strategies for AD. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Elisa Conti
- Laboratory of Neurobiology, School of Medicine and Milan Center for Neuroscience, University of Milano-Bicocca, Monza, Italy
| | - Lucio Tremolizzo
- Laboratory of Neurobiology, School of Medicine and Milan Center for Neuroscience, University of Milano-Bicocca, Monza, Italy.,Neurology, San Gerardo Hospital, Monza, Italy
| | - Marta Elena Santarone
- Laboratory of Neurobiology, School of Medicine and Milan Center for Neuroscience, University of Milano-Bicocca, Monza, Italy
| | - Marco Tironi
- Laboratory of Neurobiology, School of Medicine and Milan Center for Neuroscience, University of Milano-Bicocca, Monza, Italy
| | - Isabella Radice
- Laboratory of Neurobiology, School of Medicine and Milan Center for Neuroscience, University of Milano-Bicocca, Monza, Italy
| | - Chiara Paola Zoia
- Laboratory of Neurobiology, School of Medicine and Milan Center for Neuroscience, University of Milano-Bicocca, Monza, Italy
| | | | | | - Roberto Dominici
- Laboratory of Chemical and Clinical Analyses, Magenta Hospital, Italy
| | - Marco Casati
- Laboratory of Chemical and Clinical Analyses, San Gerardo Hospital, Monza, Italy
| | - Ildebrando Appollonio
- Laboratory of Neurobiology, School of Medicine and Milan Center for Neuroscience, University of Milano-Bicocca, Monza, Italy.,Neurology, San Gerardo Hospital, Monza, Italy
| | - Carlo Ferrarese
- Laboratory of Neurobiology, School of Medicine and Milan Center for Neuroscience, University of Milano-Bicocca, Monza, Italy.,Neurology, San Gerardo Hospital, Monza, Italy
| |
Collapse
|
34
|
Belkouch M, Hachem M, Elgot A, Lo Van A, Picq M, Guichardant M, Lagarde M, Bernoud-Hubac N. The pleiotropic effects of omega-3 docosahexaenoic acid on the hallmarks of Alzheimer's disease. J Nutr Biochem 2016; 38:1-11. [PMID: 27825512 DOI: 10.1016/j.jnutbio.2016.03.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/14/2015] [Accepted: 03/03/2016] [Indexed: 12/17/2022]
Abstract
Among omega-3 polyunsaturated fatty acids (PUFAs), docosahexaenoic acid (DHA, 22:6n-3) is important for adequate brain development and cognition. DHA is highly concentrated in the brain and plays an essential role in brain functioning. DHA, one of the major constituents in fish fats, readily crosses the blood-brain barrier from blood to the brain. Its critical role was further supported by its reduced levels in the brain of Alzheimer's disease (AD) patients. This agrees with a potential role of DHA in memory, learning and cognitive processes. Since there is yet no cure for dementia such as AD, there is growing interest in the role of DHA-supplemented diet in the prevention of AD pathogenesis. Accordingly, animal, epidemiological, preclinical and clinical studies indicated that DHA has neuroprotective effects in a number of neurodegenerative conditions including AD. The beneficial effects of this key omega-3 fatty acid supplementation may depend on the stage of disease progression, other dietary mediators and the apolipoprotein ApoE genotype. Herein, our review investigates, from animal and cell culture studies, the molecular mechanisms involved in the neuroprotective potential of DHA with emphasis on AD.
Collapse
Affiliation(s)
- Mounir Belkouch
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France.
| | - Mayssa Hachem
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Abdeljalil Elgot
- Laboratoire des Sciences et Technologies de la Santé, Unité des Sciences Biomédicales, Institut Supérieur des Sciences de la Santé, Université Hassan 1er, Settat, Morocco
| | - Amanda Lo Van
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Madeleine Picq
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Michel Guichardant
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Michel Lagarde
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Nathalie Bernoud-Hubac
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| |
Collapse
|
35
|
Park JC, Ma J, Jeon WK, Han JS. Fructus mume extracts alleviate cognitive impairments in 5XFAD transgenic mice. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 16:54. [PMID: 26852239 PMCID: PMC4744392 DOI: 10.1186/s12906-016-1033-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 02/01/2016] [Indexed: 01/01/2023]
Abstract
Background Fructus mume (F. mume) has been used as a traditional treatment for ulcer, cough, and digestive problems for many years in Asian countries. Previous studies have demonstrated that F. mume extracts alleviate cognitive deficits in rats with chronic cerebral hypoperfusion and in mice with scopolamine treatments. The present experiment was conducted to examine the effects of F. mume on cognitive impairments in 5XFAD transgenic mice with five familial Alzheimer’s disease (AD) mutations. Methods F. mume was administered daily to 5XFAD mice at 12 weeks of age and continued for 90 days. Cognitive function was evaluated using a spatial memory version of the Morris water maze task, the object/location novelty recognition test, and contextual fear conditioning at 24 weeks of age. To elucidate the possible mechanisms underlying the memory improving effects of F. mume in 5XFAD mice, we examined alterations in hippocampal cholinergic function. Results Vehicle-treated 5XFAD mice exhibited hippocampus-dependent memory impairments compared with non-transgenic littermates, which was reversed in F. mume-treated 5XFAD mice. In addition, reduced hippocampal choline acetyltransferase (ChAT) levels in 5XFAD mice were reversed by F. mume treatment, indicating that F. mume enhances the effects of cholinergic neuronal function. Conclusions F. mume may have therapeutic effects on cognitive impairments in AD.
Collapse
|
36
|
Choong XY, Tosh JL, Pulford LJ, Fisher EMC. Dissecting Alzheimer disease in Down syndrome using mouse models. Front Behav Neurosci 2015; 9:268. [PMID: 26528151 PMCID: PMC4602094 DOI: 10.3389/fnbeh.2015.00268] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 09/21/2015] [Indexed: 11/13/2022] Open
Abstract
Down syndrome (DS) is a common genetic condition caused by the presence of three copies of chromosome 21 (trisomy 21). This greatly increases the risk of Alzheimer disease (AD), but although virtually all people with DS have AD neuropathology by 40 years of age, not all develop dementia. To dissect the genetic contribution of trisomy 21 to DS phenotypes including those relevant to AD, a range of DS mouse models has been generated which are trisomic for chromosome segments syntenic to human chromosome 21. Here, we consider key characteristics of human AD in DS (AD-DS), and our current state of knowledge on related phenotypes in AD and DS mouse models. We go on to review important features needed in future models of AD-DS, to understand this type of dementia and so highlight pathogenic mechanisms relevant to all populations at risk of AD.
Collapse
Affiliation(s)
- Xun Yu Choong
- Department of Neurodegenerative Disease, Institute of Neurology, University College London London, UK ; The LonDownS Consortium London, UK
| | - Justin L Tosh
- Department of Neurodegenerative Disease, Institute of Neurology, University College London London, UK ; The LonDownS Consortium London, UK
| | - Laura J Pulford
- Department of Neurodegenerative Disease, Institute of Neurology, University College London London, UK ; The LonDownS Consortium London, UK
| | - Elizabeth M C Fisher
- Department of Neurodegenerative Disease, Institute of Neurology, University College London London, UK ; The LonDownS Consortium London, UK
| |
Collapse
|
37
|
Zou C, Montagna E, Shi Y, Peters F, Blazquez-Llorca L, Shi S, Filser S, Dorostkar MM, Herms J. Intraneuronal APP and extracellular Aβ independently cause dendritic spine pathology in transgenic mouse models of Alzheimer's disease. Acta Neuropathol 2015; 129:909-20. [PMID: 25862638 PMCID: PMC4436699 DOI: 10.1007/s00401-015-1421-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 04/01/2015] [Accepted: 04/01/2015] [Indexed: 12/28/2022]
Abstract
Alzheimer’s disease (AD) is thought to be caused by accumulation of amyloid-β protein (Aβ), which is a cleavage product of amyloid precursor protein (APP). Transgenic mice overexpressing APP have been used to recapitulate amyloid-β pathology. Among them, APP23 and APPswe/PS1deltaE9 (deltaE9) mice are extensively studied. APP23 mice express APP with Swedish mutation and develop amyloid plaques late in their life, while cognitive deficits are observed in young age. In contrast, deltaE9 mice with mutant APP and mutant presenilin-1 develop amyloid plaques early but show typical cognitive deficits in old age. To unveil the reasons for different progressions of cognitive decline in these commonly used mouse models, we analyzed the number and turnover of dendritic spines as important structural correlates for learning and memory. Chronic in vivo two-photon imaging in apical tufts of layer V pyramidal neurons revealed a decreased spine density in 4–5-month-old APP23 mice. In age-matched deltaE9 mice, in contrast, spine loss was only observed on cortical dendrites that were in close proximity to amyloid plaques. In both cases, the reduced spine density was caused by decreased spine formation. Interestingly, the patterns of alterations in spine morphology differed between these two transgenic mouse models. Moreover, in APP23 mice, APP was found to accumulate intracellularly and its content was inversely correlated with the absolute spine density and the relative number of mushroom spines. Collectively, our results suggest that different pathological mechanisms, namely an intracellular accumulation of APP or extracellular amyloid plaques, may lead to spine abnormalities in young adult APP23 and deltaE9 mice, respectively. These distinct features, which may represent very different mechanisms of synaptic failure in AD, have to be taken into consideration when translating results from animal studies to the human disease.
Collapse
|
38
|
Limited effects of an eIF2αS51A allele on neurological impairments in the 5xFAD mouse model of Alzheimer's disease. Neural Plast 2015; 2015:825157. [PMID: 25883808 PMCID: PMC4391319 DOI: 10.1155/2015/825157] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 02/15/2015] [Accepted: 02/23/2015] [Indexed: 02/02/2023] Open
Abstract
Alzheimer's disease (AD) has been associated with increased phosphorylation of the translation initiation factor 2α (eIF2α) at serine 51. Increased phosphorylation of eIF2α alters translational control and may thereby have adverse effects on synaptic plasticity, learning, and memory. To analyze if increased levels of p-eIF2α indeed promote AD-related neurocognitive impairments, we crossed 5xFAD transgenic mice with an eIF2αS51A knock-in line that expresses the nonphosphorylatable eIF2α variant eIF2αS51A. Behavioral assessment of the resulting mice revealed motor and cognitive deficits in 5xFAD mice that were, with the possible exception of locomotor hyperactivity, not restored by the eIF2αS51A allele. Telemetric intracranial EEG recordings revealed no measurable effects of the eIF2αS51A allele on 5xFAD-associated epileptic activity. Microarray-based transcriptome analyses showed clear transcriptional alterations in 5xFAD hippocampus that were not corrected by the eIF2αS51A allele. In contrast to prior studies, our immunoblot analyses did not reveal increased levels of p-eIF2α in the hippocampus of 5xFAD mice, suggesting that elevated p-eIF2α levels are not a universal feature of AD models. Collectively, our data indicate that 5xFAD-related pathologies do not necessarily require hyperphosphorylation of eIF2α to emerge; they also show that heterozygosity for the nonphosphorylatable eIF2αS51A allele has limited effects on 5xFAD-related disease manifestations.
Collapse
|
39
|
Jadalannagari S, Aljitawi OS. Ectodermal Differentiation of Wharton's Jelly Mesenchymal Stem Cells for Tissue Engineering and Regenerative Medicine Applications. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:314-22. [PMID: 25517045 DOI: 10.1089/ten.teb.2014.0404] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSCs) from Wharton's jelly (WJ) of the human umbilical cord are perinatal stem cells that have self-renewal ability, extended proliferation potential, immunosuppressive properties, and are accordingly excellent candidates for tissue engineering. These MSCs are unique, easily accessible, and a noncontroversial cell source of regeneration in medicine. Wharton's jelly mesenchymal stem cells (WJMSCs) are multipotent and capable of multilineage differentiation into cells like adipocytes, bone, cartilage, and skeletal muscle upon exposure to appropriate conditions. The ectoderm is one of the three primary germ layers found in the very early embryo that differentiates into the epidermis, nervous system (spine, peripheral nerves, brain), and exocrine glands (mammary, sweat, salivary, and lacrimal glands). Accumulating evidence shows that MSCs obtained from WJ have an ectodermal differentiation potential. The current review examines this differentiation potential of WJMSC into the hair follicle, skin, neurons, and sweat glands along with discussing the potential utilization of such differentiation in regenerative medicine.
Collapse
Affiliation(s)
| | - Omar S Aljitawi
- 1Department of Bioengineering, University of Kansas, Lawrence, Kansas.,2Department of Hematology/Oncology, Blood and Marrow Transplant Program, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
40
|
Grothe MJ, Ewers M, Krause B, Heinsen H, Teipel SJ. Basal forebrain atrophy and cortical amyloid deposition in nondemented elderly subjects. Alzheimers Dement 2014; 10:S344-53. [PMID: 24418052 PMCID: PMC4092050 DOI: 10.1016/j.jalz.2013.09.011] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 08/09/2013] [Accepted: 09/04/2013] [Indexed: 02/05/2023]
Abstract
BACKGROUND Both neurodegeneration of the cholinergic basal forebrain (BF) and deposition of β-amyloid are early events in the course of Alzheimer's disease (AD). Associations between increased amyloid pathology and cholinergic atrophy have been described in autopsy studies. METHODS We used structural MRI and AV45-PET amyloid imaging data of 225 cognitively normal or mildly impaired elderly subjects from the Alzheimer's Disease Neuroimaging Initiative to assess in vivo associations between BF atrophy and cortical amyloid deposition. Associations were examined using region-of-interest (ROI) and voxel-based approaches with reference to cytoarchitectonic mappings of the cholinergic BF nuclei. RESULTS ROI- and voxel-based approaches yielded complementary evidence for an association between BF volume and cortical amyloid deposition in presymptomatic and predementia stages of AD, irrespective of age, gender, and APOE genotype. CONCLUSIONS The observed correlations between BF atrophy and cortical amyloid load likely reflect associations between cholinergic degeneration and amyloid pathology as reported in neuropathologic examination studies.
Collapse
Affiliation(s)
- Michel J Grothe
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany.
| | - Michael Ewers
- Institute for Stroke and Dementia Research, Klinikum Grosshadern, Ludwig-Maximilians University, Munich, Germany
| | - Bernd Krause
- Department of Nuclear Medicine, University of Rostock, Rostock, Germany
| | - Helmut Heinsen
- Laboratory of Morphological Brain Research, Department of Psychiatry, University of Würzburg, Würtzburg, Germany
| | - Stefan J Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany; Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| |
Collapse
|
41
|
|
42
|
Parikh V, Bernard CS, Naughton SX, Yegla B. Interactions between Aβ oligomers and presynaptic cholinergic signaling: age-dependent effects on attentional capacities. Behav Brain Res 2014; 274:30-42. [PMID: 25101540 DOI: 10.1016/j.bbr.2014.07.046] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 07/14/2014] [Accepted: 07/25/2014] [Indexed: 12/24/2022]
Abstract
Substantial evidence suggests that cerebral deposition of the neurotoxic fibrillar form of amyloid precursor protein, β-amyloid (Aβ), plays a critical role in the pathogenesis of Alzheimer's disease (AD). Yet, many aspects of AD pathology including the cognitive symptoms and selective vulnerability of cortically projecting basal forebrain (BF) cholinergic neurons are not well explained by this hypothesis. Specifically, it is not clear why cognitive decline appears early when the loss of BF cholinergic neurons and plaque deposition are manifested late in AD. Soluble oligomeric forms of Aβ are proposed to appear early in the pathology and to be better predictors of synaptic loss and cognitive deficits. The present study was designed to examine the impact of Aβ oligomers on attentional functions and presynaptic cholinergic transmission in young and aged rats. Chronic intracranial infusions of Aβ oligomers produced subtle decrements in the ability of rats to sustain attentional performance with time on task, irrespective of the age of the animals. However, Aβ oligomers produced robust detrimental effects on performance under conditions of enhanced attentional load in aged animals. In vivo electrochemical recordings show reduced depolarization-evoked cholinergic signals in Aβ-infused aged rats. Moreover, soluble Aβ disrupted the capacity of cholinergic synapses to clear exogenous choline from the extracellular space in both young and aged rats, reflecting impairments in the choline transport process that is critical for acetylcholine (ACh) synthesis and release. Although aging per se reduced the cross-sectional area of BF cholinergic neurons and presynaptic cholinergic proteins in the cortex, attentional performance and ACh release remained unaffected in aged rats infused with the control peptide. Taken together, these data suggest that soluble Aβ may marginally influence attentional functions at young ages primarily by interfering with the choline uptake processes. However, age-related weakening of the cholinergic system may synergistically interact with these disruptive presynaptic mechanisms to make this neurotransmitter system vulnerable to the toxic effects of oligomeric Aβ in robustly impeding attentional capacities.
Collapse
Affiliation(s)
- Vinay Parikh
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, United States.
| | - Carcha S Bernard
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, United States
| | - Sean X Naughton
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, United States
| | - Brittney Yegla
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, United States
| |
Collapse
|
43
|
Chronic neuroinflammation in Alzheimer's disease: new perspectives on animal models and promising candidate drugs. BIOMED RESEARCH INTERNATIONAL 2014; 2014:309129. [PMID: 25025046 PMCID: PMC4083880 DOI: 10.1155/2014/309129] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/14/2014] [Accepted: 05/14/2014] [Indexed: 12/03/2022]
Abstract
Chronic neuroinflammation is now considered one of the major factors in the pathogenesis of Alzheimer's disease (AD). However, the most widely used transgenic AD models (overexpressing mutated forms of amyloid precursor protein, presenilin, and/or tau) do not demonstrate the degree of inflammation, neurodegeneration (particularly of the cholinergic system), and cognitive decline that is comparable with the human disease. Hence a more suitable animal model is needed to more closely mimic the resulting cognitive decline and memory loss in humans in order to investigate the effects of neuroinflammation on neurodegeneration. One of these models is the glial fibrillary acidic protein-interleukin 6 (GFAP-IL6) mouse, in which chronic neuroinflammation triggered constitutive expression of the cytokine interleukin-6 (IL-6) in astrocytes. These transgenic mice show substantial and progressive neurodegeneration as well as a decline in motor skills and cognitive function, starting from 6 months of age. This animal model could serve as an excellent tool for drug discovery and validation in vivo. In this review, we have also selected three potential anti-inflammatory drugs, curcumin, apigenin, and tenilsetam, as candidate drugs, which could be tested in this model.
Collapse
|
44
|
Protective effects of Borago officinalis extract on amyloid β-peptide(25-35)-induced memory impairment in male rats: a behavioral study. BIOMED RESEARCH INTERNATIONAL 2014; 2014:798535. [PMID: 25013802 PMCID: PMC4071970 DOI: 10.1155/2014/798535] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/19/2014] [Accepted: 05/21/2014] [Indexed: 01/28/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder and most common form of dementia that leads to memory impairment. In the present study we have examined the protective effects of Borago officinalis (borage) extract on Amyloid β (A β)-Induced memory impairment. Wistar male rats received intrahippocampal (IHP) injection of the A β (25-35) and borage extract throughout gestation (100 mg/kg). Learning and memory functions in the rats were examined by the passive avoidance and the Morris water maze (MWM) tasks. Finally, the antioxidant capacity of hippocampus was measured using ferric ion reducing antioxidant power (FRAP) assay. The results showed that A β (25-35) impaired step-through latency and time in dark compartment in passive avoidance task. In the MWM, A β (25-35) significantly increased escape latency and traveled distance. Borage administration attenuated the A β-induced memory impairment in both the passive avoidance and the MWM tasks. A β induced a remarkable decrease in antioxidant power (FRAP value) of hippocampus and borage prevented the decrease of the hippocampal antioxidant status. This data suggests that borage could improve the learning impairment and oxidative damage in the hippocampal tissue following A β treatment and that borage consumption may lead to an improvement of AD-induced cognitive dysfunction.
Collapse
|
45
|
Abstract
The basal forebrain cholinergic system modulates neuronal excitability and vascular tone throughout the cerebral cortex and hippocampus. This system is severely affected in Alzheimer's disease (AD), and drug treatment to enhance cholinergic signaling is widely used as symptomatic therapy in AD. Defining the full morphologies of individual basal forebrain cholinergic neurons has, until now, been technically beyond reach due to their large axon arbor sizes. Using genetically-directed sparse labeling, we have characterized the complete morphologies of basal forebrain cholinergic neurons in the mouse. Individual arbors were observed to span multiple cortical columns, and to have >1000 branch points and total axon lengths up to 50 cm. In an AD model, cholinergic axons were slowly lost and there was an accumulation of axon-derived material in discrete puncta. Calculations based on published morphometric data indicate that basal forebrain cholinergic neurons in humans have a mean axon length of ∼100 meters. DOI:http://dx.doi.org/10.7554/eLife.02444.001 The human brain is made up of roughly 80 to 100 billion neurons, organized into extensive networks. Each neuron consists of a number of components: a cell body, which contains the nucleus; numerous short protrusions from the cell body called dendrites; and a long thin structure called an axon that carries the electrical signals generated in the cell body and the dendrites to the next neuron in the network. One of the most studied networks in the human brain is the basal forebrain network, which is made up of large neurons that communicate with one another using a chemical transmitter called acetylcholine. This network has a key role in cognition, and its neurons are among the first to degenerate in Alzheimer's disease. However, relatively little is known about the structure of these ‘cholinergic’ neurons because their large size makes them difficult to study using standard techniques. Now, Wu et al. have visualized, for the first time, the complete 3D structure of cholinergic neurons in the mouse forebrain. The mice in question had been genetically modified so that only ten or so of their many thousands of cholinergic neurons expressed a distinctive ‘marker’ protein. This made it possible to distinguish these neurons from surrounding brain tissue in order to visualize their structures. The resulting pictures clearly illustrate the neurons' complexity, with individual axons in adult mice displaying up to 1000 branches. Measurements showed that each cholinergic axon in the mouse brain is roughly 30 centimeters long, even though the brain itself is less than 2 centimeters from front to back. Based on measurements by other researchers, Wu et al. calculated that the axons of single cholinergic neurons in the human brain are about 100 meters long on average. The extreme length and complex branching structure of cholinergic forebrain neurons helps to explain why each neuron is able to modulate the activity of many others in the network. It could also explain their vulnerability to degeneration, as the need to transport materials over such long distances may limit the ability of these neurons to respond to damage. DOI:http://dx.doi.org/10.7554/eLife.02444.002
Collapse
Affiliation(s)
- Hao Wu
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, United States
| | - John Williams
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Jeremy Nathans
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, United States Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States Department of Opthalmology, Johns Hopkins University School of Medicine, Baltimore, United States
| |
Collapse
|
46
|
Bilkei-Gorzo A. Genetic mouse models of brain ageing and Alzheimer's disease. Pharmacol Ther 2014; 142:244-57. [DOI: 10.1016/j.pharmthera.2013.12.009] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 11/26/2013] [Indexed: 12/21/2022]
|
47
|
Härtig W, Saul A, Kacza J, Grosche J, Goldhammer S, Michalski D, Wirths O. Immunolesion-induced loss of cholinergic projection neurones promotes β-amyloidosis and tau hyperphosphorylation in the hippocampus of triple-transgenic mice. Neuropathol Appl Neurobiol 2014; 40:106-20. [DOI: 10.1111/nan.12050] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 03/28/2013] [Indexed: 01/13/2023]
Affiliation(s)
- W. Härtig
- Paul Flechsig Institute for Brain Research; University of Leipzig; Leipzig Germany
| | - A. Saul
- Division of Molecular Psychiatry; Department of Psychiatry; University of Göttingen; Göttingen Germany
| | - J. Kacza
- Institute of Anatomy, Histology and Embryology; Faculty of Veterinary Medicine; University of Leipzig; Leipzig Germany
| | - J. Grosche
- Paul Flechsig Institute for Brain Research; University of Leipzig; Leipzig Germany
| | - S. Goldhammer
- Paul Flechsig Institute for Brain Research; University of Leipzig; Leipzig Germany
| | - D. Michalski
- Department of Neurology; University of Leipzig; Leipzig Germany
| | - O. Wirths
- Division of Molecular Psychiatry; Department of Psychiatry; University of Göttingen; Göttingen Germany
| |
Collapse
|
48
|
Gratwicke J, Kahan J, Zrinzo L, Hariz M, Limousin P, Foltynie T, Jahanshahi M. The nucleus basalis of Meynert: A new target for deep brain stimulation in dementia? Neurosci Biobehav Rev 2013; 37:2676-88. [DOI: 10.1016/j.neubiorev.2013.09.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 08/30/2013] [Accepted: 09/02/2013] [Indexed: 10/26/2022]
|
49
|
Teipel S, Heinsen H, Amaro E, Grinberg LT, Krause B, Grothe M. Cholinergic basal forebrain atrophy predicts amyloid burden in Alzheimer's disease. Neurobiol Aging 2013; 35:482-91. [PMID: 24176625 DOI: 10.1016/j.neurobiolaging.2013.09.029] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 09/09/2013] [Accepted: 09/19/2013] [Indexed: 01/24/2023]
Abstract
We compared accuracy of hippocampus and basal forebrain cholinergic system (BFCS) atrophy to predict cortical amyloid burden in 179 cognitively normal subjects (CN), 269 subjects with early stages of mild cognitive impairment (MCI), 136 subjects with late stages of MCI, and 86 subjects with Alzheimer's disease (AD) dementia retrieved from the Alzheimer's Disease Neuroimaging Initiative database. Hippocampus and BFCS volumes were determined from structural magnetic resonance imaging scans at 3 Tesla, and cortical amyloid load from AV45 (florbetapir) positron emission tomography scans. In receiver operating characteristics analyses, BFCS volume provided significantly more accurate classification into amyloid-negative and -positive categories than hippocampus volume. In contrast, hippocampus volume more accurately identified the diagnostic categories of AD, late and early MCI, and CN compared with whole and anterior BFCS volume, whereas posterior BFCS and hippocampus volumes yielded similar diagnostic accuracy. In logistic regression analysis, hippocampus and posterior BFCS volumes contributed significantly to discriminate MCI and AD from CN, but only BFCS volume predicted amyloid status. Our findings suggest that BFCS atrophy is more closely associated with cortical amyloid burden than hippocampus atrophy in predementia AD.
Collapse
Affiliation(s)
- Stefan Teipel
- Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany; DZNE, German Center for Neurodegenerative Disorders, Rostock, Germany.
| | | | | | | | | | | | | |
Collapse
|
50
|
Zhao L, Chu CB, Li JF, Yang YT, Niu SQ, Qin W, Hao YG, Dong Q, Guan R, Hu WL, Wang Y. Glycogen synthase kinase-3 reduces acetylcholine level in striatum via disturbing cellular distribution of choline acetyltransferase in cholinergic interneurons in rats. Neuroscience 2013; 255:203-11. [PMID: 24121130 DOI: 10.1016/j.neuroscience.2013.10.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 09/12/2013] [Accepted: 10/01/2013] [Indexed: 12/31/2022]
Abstract
Cholinergic interneurons, which provide the main source of acetylcholine (ACh) in the striatum, control the striatal local circuits and deeply involve in the pathogenesis of neurodegenerative diseases. Glycogen synthase kinase-3 (GSK-3) is a crucial kinase with diverse fundamental functions and accepted that deregulation of GSK-3 activity also plays important roles in diverse neurodegenerative diseases. However, up to now, there is no direct proof indicating whether GSK-3 activation is responsible for cholinergic dysfunction. In the present study, with combined intracerebroventricular injection of Wortmannin and GF-109203X, we activated GSK-3 and demonstrated the increased phosphorylation level of microtubule-associated protein tau and neurofilaments (NFs) in the rat striatum. The activated GSK-3 consequently decreased ACh level in the striatum as a result of the reduction of choline acetyltransferase (ChAT) activity. The alteration of ChAT activity was due to impaired ChAT distribution rather than its expression. Furthermore, we proved that cellular ChAT distribution was dependent on low phosphorylation level of NFs. Nevertheless, the cholinergic dysfunction in the striatum failed to induce significant neuronal number reduction. In summary, our data demonstrates the link between GSK-3 activation and cholinergic dysfunction in the striatum and provided beneficial evidence for the pathogenesis study of relevant neurodegenerative diseases.
Collapse
Affiliation(s)
- L Zhao
- Department of Neurobiology and Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|