1
|
Vockert N, Machts J, Kleineidam L, Nemali A, Incesoy EI, Bernal J, Schütze H, Yakupov R, Peters O, Gref D, Schneider LS, Preis L, Priller J, Spruth EJ, Altenstein S, Schneider A, Fliessbach K, Wiltfang J, Rostamzadeh A, Glanz W, Teipel S, Kilimann I, Goerss D, Laske C, Munk MH, Spottke A, Roy N, Heneka MT, Brosseron F, Wagner M, Wolfsgruber S, Dobisch L, Dechent P, Hetzer S, Scheffler K, Zeidman P, Stern Y, Schott BH, Jessen F, Düzel E, Maass A, Ziegler G. Cognitive reserve against Alzheimer's pathology is linked to brain activity during memory formation. Nat Commun 2024; 15:9815. [PMID: 39537609 PMCID: PMC11561234 DOI: 10.1038/s41467-024-53360-9] [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: 10/30/2023] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
The cognitive reserve (CR) hypothesis posits that individuals can differ in how their brain function is disrupted by pathology associated with aging and neurodegeneration. Here, we test this hypothesis in the continuum from cognitively normal to at-risk stages for Alzheimer's Disease (AD) to AD dementia using longitudinal data from 490 participants of the DELCODE multicentric observational study. Brain function is measured using task fMRI of visual memory encoding. Using a multivariate moderation analysis, we identify a CR-related activity pattern underlying successful memory encoding that moderates the detrimental effect of AD pathological load on cognitive performance. CR is mainly represented by a more pronounced expression of the task-active network encompassing deactivation of the default mode network (DMN) and activation of inferior temporal regions including the fusiform gyrus. We devise personalized fMRI-based CR scores that moderate the impact of AD pathology on cognitive performance and are positively associated with years of education. Furthermore, higher CR scores attenuate the effect of AD pathology on cognitive decline over time. Our findings primarily provide evidence for the maintenance of core cognitive circuits including the DMN as the neural basis of CR. Individual brain activity levels of these areas during memory encoding have prognostic value for future cognitive decline.
Collapse
Affiliation(s)
- Niklas Vockert
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.
| | - Judith Machts
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Department of Neurodegenerative Diseases and Geriatric Psychiatry, Bonn, Germany
| | - Aditya Nemali
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Enise I Incesoy
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
- Department for Psychiatry and Psychotherapy, University Clinic Magdeburg, Magdeburg, Germany
| | - Jose Bernal
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Hartmut Schütze
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Renat Yakupov
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Charité - Universitaetsmedizin Berlin, corporate member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Daria Gref
- Charité - Universitaetsmedizin Berlin, corporate member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Luisa Sophie Schneider
- Charité - Universitaetsmedizin Berlin, corporate member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, ECRC Experimental and Clinical Research Center, Berlin, Germany
| | - Lukas Preis
- Charité - Universitaetsmedizin Berlin, corporate member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité Berlin, Germany
- School of Medicine, Technical University of Munich, Department of Psychiatry and Psychotherapy, Munich, Germany
- University of Edinburgh and UK DRI, Edinburgh, UK
| | - Eike Jakob Spruth
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité Berlin, Germany
| | - Slawek Altenstein
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité Berlin, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Department of Neurodegenerative Diseases and Geriatric Psychiatry, Bonn, Germany
| | - Klaus Fliessbach
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Department of Neurodegenerative Diseases and Geriatric Psychiatry, Bonn, Germany
| | - Jens Wiltfang
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Goettingen, Germany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Ayda Rostamzadeh
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Doreen Goerss
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, 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 Tuebingen, Tuebingen, Germany
| | - Matthias H Munk
- German Center for Neurodegenerative Diseases (DZNE), Tuebingen, Germany
- Department of Psychiatry and Psychotherapy, University of Tuebingen, Tuebingen, Germany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, University of Bonn, Bonn, Germany
| | - Nina Roy
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Michael T Heneka
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Luxembourg, Luxembourg
| | | | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Department of Neurodegenerative Diseases and Geriatric Psychiatry, Bonn, Germany
| | - Steffen Wolfsgruber
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Department of Neurodegenerative Diseases and Geriatric Psychiatry, Bonn, Germany
| | - Laura Dobisch
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Peter Dechent
- MR-Research in Neurosciences, Department of Cognitive Neurology, Georg-August-University Goettingen, Goettingen, Germany
| | - Stefan Hetzer
- Berlin Center for Advanced Neuroimaging, Charité - Universitaetsmedizin Berlin, Berlin, Germany
| | - Klaus Scheffler
- Department for Biomedical Magnetic Resonance, University of Tuebingen, Tuebingen, Germany
| | - Peter Zeidman
- Wellcome Centre for Human Neuroimaging, UCL Institute of Neurology, London, UK
| | - Yaakov Stern
- Cognitive Neuroscience Division, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Björn H Schott
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Goettingen, Germany
- Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Psychiatry, University of Cologne, Koeln, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Koeln, 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
| | - Anne Maass
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.
| | - Gabriel Ziegler
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
2
|
Gong M, Fang Y, Yang K, Yuan F, Hu R, Su Y, Yang Y, Xu W, Ma Q, Cha J, Zhang R, Zhang Z, Li W. The WFS1-ZnT3-Zn 2+ Axis Regulates the Vicious Cycle of Obesity and Depression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403405. [PMID: 39258564 PMCID: PMC11538679 DOI: 10.1002/advs.202403405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/20/2024] [Indexed: 09/12/2024]
Abstract
Obesity, a growing global health concern, is closely linked to depression. However, the neural mechanism of association between obesity and depression remains poorly understood. In this study, neural-specific WFS1 deficiency exacerbates the vicious cycle of obesity and depression in mice fed a high-fat diet (HFD), positioning WFS1 as a crucial factor in this cycle. Through human pluripotent stem cells (hESCs) neural differentiation, it is demonstrated that WFS1 regulates Zn2+ homeostasis and the apoptosis of neural progenitor cells (NPCs) and cerebral organoids by inhibiting the zinc transporter ZnT3 under the situation of dysregulated lipid metabolism. Notably, riluzole regulates ZnT3 expression to maintain zinc homeostasis and protect NPCs from lipotoxicity-induced cell death. Importantly, riluzole, a therapeutic molecule targeting the nervous system, in vivo administration prevents HFD-induced obesity and associated depression. Thus, a WFS1-ZnT3-Zn2+ axis critical is demonstrated for the vicious cycle of obesity and depression and that riluzole may have the potential to reverse this process against obesity and depression.
Collapse
Affiliation(s)
- Mengting Gong
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Yulin Fang
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Kaijiang Yang
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Fei Yuan
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Rui Hu
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Yajuan Su
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Yiling Yang
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Wenjun Xu
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Qing Ma
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Jiaxue Cha
- Shanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Ru Zhang
- Shanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Zhen‐Ning Zhang
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Weida Li
- Institute for Regenerative MedicineState Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| |
Collapse
|
3
|
Caruso V, Rigoli L. Beyond Wolfram Syndrome 1: The WFS1 Gene's Role in Alzheimer's Disease and Sleep Disorders. Biomolecules 2024; 14:1389. [PMID: 39595565 PMCID: PMC11591713 DOI: 10.3390/biom14111389] [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: 09/27/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
The WFS1 gene was first identified in Wolfram Syndrome 1 (WS1), a rare autosomal recessive genetic disorder characterized by severe and progressive neurodegenerative changes. WFS1's role in various cellular mechanisms, particularly in calcium homeostasis and the modulation of endoplasmic reticulum (ER) stress, suggests its potential involvement in the pathogenesis of Alzheimer's disease (AD) and sleep disorders. Because it is involved in maintaining ER balance, calcium signaling, and stress responses, WFS1 plays a multifaceted role in neuronal health. Numerous studies have shown that the absence or improper expression of WFS1 disrupts these cellular processes, leading to neurodegeneration and making neurons more vulnerable. In AD, WFS1 dysfunction is thought to contribute to the accumulation of amyloid-β (Aβ) plaques and tau tangles, thereby accelerating disease progression. Additionally, WFS1 plays an essential role in sleep regulation by influencing neuronal excitability and neurotransmitter release, which may explain the sleep disturbances frequently observed in neurodegenerative diseases. Due to its involvement in the pathological mechanisms of AD and sleep disorders, WFS1 is regarded as a potential early diagnostic marker for these diseases. Further research is required to fully elucidate WFS1's role in the cellular pathway, perhaps facilitating the development of new therapeutic strategies for Alzheimer's disease and sleep disorders.
Collapse
Affiliation(s)
- Valerio Caruso
- Department of Neuroscience, Psychiatric Section, Azienda Ospedaliera Universitaria Pisana (A.U.O.P.), 56126 Pisa, Italy;
| | - Luciana Rigoli
- Department of Human Pathology of Adulthood and Childhood G. Barresi, University of Messina, 98125 Messina, Italy
| |
Collapse
|
4
|
Zhang S, Crossley CA, Yuan Q. Neuronal Vulnerability of the Entorhinal Cortex to Tau Pathology in Alzheimer's Disease. Br J Biomed Sci 2024; 81:13169. [PMID: 39435008 PMCID: PMC11491395 DOI: 10.3389/bjbs.2024.13169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024]
Abstract
This review delves into the entorhinal cortex (EC) as a central player in the pathogenesis of Alzheimer's Disease (AD), emphasizing its role in the accumulation and propagation of tau pathology. It elucidates the multifaceted functions of the EC, encompassing memory formation, spatial navigation, and olfactory processing, while exploring how disruptions in these processes contribute to cognitive decline in AD. The review discusses the intricate interplay between tau pathology and EC vulnerability, highlighting how alterations in neuronal firing patterns and synaptic function within the EC exacerbate cognitive impairments. Furthermore, it elucidates how specific neuronal subtypes within the EC exhibit differential susceptibility to tau-induced damage, contributing to disease progression. Early detection methods, such as imaging techniques and assessments of EC blood flow, are examined as potential tools for identifying tau pathology in the preclinical stages of AD. These approaches offer promise for improving diagnostic accuracy and enabling timely intervention. Therapeutic strategies targeting tau pathology within the EC are explored, including the clearance of pathological tau aggregates and the inhibition of tau aggregation processes. By understanding the molecular and cellular mechanisms underlying EC vulnerability, researchers can develop more targeted and effective interventions to slow disease progression. The review underscores the importance of reliable biomarkers to assess disease progression and therapeutic efficacy in clinical trials targeting the EC. Ultimately, it aims to contribute to the development of more effective management strategies for AD, emphasizing the translation of research findings into clinical practice to address the growing societal burden of the disease.
Collapse
Affiliation(s)
| | - Chelsea Ann Crossley
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| | | |
Collapse
|
5
|
Kim Y, Ryu SH, Hyun J, Cho YS, Jung YK. TLR2 immunotherapy suppresses neuroinflammation, tau spread, and memory loss in rTg4510 mice. Brain Behav Immun 2024; 121:291-302. [PMID: 39098437 DOI: 10.1016/j.bbi.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024] Open
Abstract
In Alzheimer's disease, chronic neuroinflammation is accompanied by amyloid and tau pathologies. Especially, aberrant microglial activation is known to precede the regional tau pathology development, but the mechanisms how microglia affect tau spread remain largely unknown. Here, we found that toll-like receptor 2 (TLR2) in microglia recognizes oligomeric tau as a pathogenic ligand and induces inflammatory responses. Knockout of TLR2 reduced tau pathology and microglial activation in rTg4510 tau transgenic mice. Treatment of oligomeric tau induced TLR2 activation and increased inflammatory responses in microglial cells. TLR2 further mediated the tau-induced microglial activation and promoted tau uptake into neurons in neuron-microglia co-culture system and in mouse hippocampus after intracranial tau injection. Importantly, treatment with anti-TLR2 monoclonal antibody Tomaralimab blocked TLR2 activation and inflammatory responses in a dose-dependent manner, and significantly reduced tau spread and memory loss in rTg4510 mice. These results suggest that TLR2 plays a crucial role in tau spread by causing aberrant microglial activation in response to pathological tau, and blocking TLR2 with immunotherapy may ameliorate tau pathogenesis in Alzheimer's disease.
Collapse
Affiliation(s)
- Youbin Kim
- Interdisciplinary Program in Neuroscience, Seoul National University, Seoul 08826, Republic of Korea; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Shin-Hyeon Ryu
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Junho Hyun
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Young-Sin Cho
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yong-Keun Jung
- Interdisciplinary Program in Neuroscience, Seoul National University, Seoul 08826, Republic of Korea; School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
6
|
Masurkar AV, Marsh K, Morgan B, Leitner D, Wisniewski T. Factors Affecting Resilience and Prevention of Alzheimer's Disease and Related Dementias. Ann Neurol 2024; 96:633-649. [PMID: 39152774 PMCID: PMC11534551 DOI: 10.1002/ana.27055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 07/27/2024] [Accepted: 07/31/2024] [Indexed: 08/19/2024]
Abstract
Alzheimer's disease (AD) is a devastating, age-associated neurodegenerative disorder and the most common cause of dementia. The clinical continuum of AD spans from preclinical disease to subjective cognitive decline, mild cognitive impairment, and dementia stages (mild, moderate, and severe). Neuropathologically, AD is defined by the accumulation of amyloid β (Aβ) into extracellular plaques in the brain parenchyma and in the cerebral vasculature, and by abnormally phosphorylated tau that accumulates intraneuronally forming neurofibrillary tangles (NFTs). Development of treatment approaches that prevent or even reduce the cognitive decline because of AD has been slow compared to other major causes of death. Recently, the United States Food and Drug Administration gave full approval to 2 different Aβ-targeting monoclonal antibodies. However, this breakthrough disease modifying approach only applies to a limited subset of patients in the AD continuum and there are stringent eligibility criteria. Furthermore, these approaches do not prevent progression of disease, because other AD-related pathologies, such as NFTs, are not directly targeted. A non-mutually exclusive alternative is to address lifestyle interventions that can help reduce the risk of AD and AD-related dementias (ADRD). It is estimated that addressing such modifiable risk factors could potentially delay up to 40% of AD/ADRD cases. In this review, we discuss some of the many modifiable risk factors that may be associated with prevention of AD/ADRD and/or increasing brain resilience, as well as other factors that may interact with these modifiable risk factors to influence AD/ADRD progression. [Color figure can be viewed at www.annalsofneurology.org] ANN NEUROL 2024;96:633-649.
Collapse
Affiliation(s)
- Arjun V. Masurkar
- Department of Neurology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
- Center for Cognitive Neurology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
| | - Karyn Marsh
- Department of Neurology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
- Center for Cognitive Neurology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
| | - Brianna Morgan
- Department of Medicine, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
| | - Dominique Leitner
- Department of Neurology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
- Center for Cognitive Neurology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
| | - Thomas Wisniewski
- Department of Neurology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
- Center for Cognitive Neurology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
- Department of Pathology, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
- Department of Psychiatry, New York University Grossman School of Medicine, 560 First Avenue, New York, NY 10016
| |
Collapse
|
7
|
Mate de Gerando A, Khasnavis A, Welikovitch LA, Bhavsar H, Glynn C, Quittot N, Perbet R, Hyman BT. Aqueous extractable nonfibrillar and sarkosyl extractable fibrillar Alzheimer's disease tau seeds have distinct properties. Acta Neuropathol Commun 2024; 12:145. [PMID: 39252090 PMCID: PMC11382398 DOI: 10.1186/s40478-024-01849-1] [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: 07/03/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024] Open
Abstract
Pathological tau fibrils in progressive supranuclear palsy, frontotemporal dementia, chronic traumatic encephalopathy, and Alzheimer's disease each have unique conformations, and post-translational modifications that correlate with unique disease characteristics. However, within Alzheimer's disease (AD), both fibrillar (sarkosyl insoluble (AD SARK tau)), and nonfibrillar (aqueous extractable high molecular weight (AD HMW tau)) preparations have been suggested to be seed-competent. We now explore if these preparations are similar or distinct in their in vivo seeding characteristics. Using an in vivo amplification and time-course paradigm we demonstrate that, for AD HMW and AD SARK tau species, the amplified material is biochemically similar to the original sample. The HMW and SARK materials also show different clearance, propagation kinetics, and propagation patterns. These data indicate the surprising co-occurrence of multiple distinct tau species within the same AD brain, supporting the idea that multiple tau conformers - both fibrillar and nonfibrillar- can impact phenotype in AD.
Collapse
Affiliation(s)
- Anastasie Mate de Gerando
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Cambridge, MA, USA
| | - Anita Khasnavis
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | | | - Harshil Bhavsar
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Calina Glynn
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Cambridge, MA, USA
| | - Noe Quittot
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Romain Perbet
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Cambridge, MA, USA.
| |
Collapse
|
8
|
Ahuja K, Vandenabeele M, Nami F, Lefevere E, Van Hoecke J, Bergmans S, Claes M, Vervliet T, Neyrinck K, Burg T, De Herdt D, Bhaskar P, Zhu Y, Looser ZJ, Loncke J, Gsell W, Plaas M, Agostinis P, Swinnen JV, Van Den Bosch L, Bultynck G, Saab AS, Wolfs E, Chai YC, Himmelreich U, Verfaillie C, Moons L, De Groef L. A deep phenotyping study in mouse and iPSC models to understand the role of oligodendroglia in optic neuropathy in Wolfram syndrome. Acta Neuropathol Commun 2024; 12:140. [PMID: 39198924 PMCID: PMC11351506 DOI: 10.1186/s40478-024-01851-7] [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: 04/29/2024] [Accepted: 07/18/2024] [Indexed: 09/01/2024] Open
Abstract
Wolfram syndrome (WS) is a rare childhood disease characterized by diabetes mellitus, diabetes insipidus, blindness, deafness, neurodegeneration and eventually early death, due to autosomal recessive mutations in the WFS1 (and WFS2) gene. While it is categorized as a neurodegenerative disease, it is increasingly becoming clear that other cell types besides neurons may be affected and contribute to the pathogenesis. MRI studies in patients and phenotyping studies in WS rodent models indicate white matter/myelin loss, implicating a role for oligodendroglia in WS-associated neurodegeneration. In this study, we sought to determine if oligodendroglia are affected in WS and whether their dysfunction may be the primary cause of the observed optic neuropathy and brain neurodegeneration. We demonstrate that 7.5-month-old Wfs1∆exon8 mice display signs of abnormal myelination and a reduced number of oligodendrocyte precursor cells (OPCs) as well as abnormal axonal conduction in the optic nerve. An MRI study of the brain furthermore revealed grey and white matter loss in the cerebellum, brainstem, and superior colliculus, as is seen in WS patients. To further dissect the role of oligodendroglia in WS, we performed a transcriptomics study of WS patient iPSC-derived OPCs and pre-myelinating oligodendrocytes. Transcriptional changes compared to isogenic control cells were found for genes with a role in ER function. However, a deep phenotyping study of these WS patient iPSC-derived oligodendroglia unveiled normal differentiation, mitochondria-associated endoplasmic reticulum (ER) membrane interactions and mitochondrial function, and no overt signs of ER stress. Overall, the current study indicates that oligodendroglia functions are largely preserved in the WS mouse and patient iPSC-derived models used in this study. These findings do not support a major defect in oligodendroglia function as the primary cause of WS, and warrant further investigation of neurons and neuron-oligodendroglia interactions as a target for future neuroprotective or -restorative treatments for WS.
Collapse
Affiliation(s)
- K Ahuja
- Cellular Communication and Neurodegeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - M Vandenabeele
- Cellular Communication and Neurodegeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - F Nami
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - E Lefevere
- Cellular Communication and Neurodegeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - J Van Hoecke
- Cellular Communication and Neurodegeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - S Bergmans
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - M Claes
- Cellular Communication and Neurodegeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - T Vervliet
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - K Neyrinck
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - T Burg
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute, KU Leuven, Leuven, Belgium
- VIB Center for Brain & Disease Research, Laboratory of Neurobiology, VIB-KU Leuven, Leuven, Belgium
| | - D De Herdt
- Cellular Communication and Neurodegeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - P Bhaskar
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Y Zhu
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Z J Looser
- Institute of Pharmacology and Toxicology, Neuroscience Center Zurich, University of Zurich, University and ETH Zurich, Zurich, Switzerland
| | - J Loncke
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - W Gsell
- Biomedical MRI Group/MoSAIC, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - M Plaas
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - P Agostinis
- Laboratory for Cell Death Research & Therapy, Department of Cellular and Molecular Medicine, Leuven Center for Cancer Biology, VIB-KU, Leuven Cancer Institute, VIB-KU Leuven, Leuven, Belgium
| | - J V Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven Institute for Single Cell Omics (LISCO), KU Leuven, Leuven, Belgium
| | - L Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute, KU Leuven, Leuven, Belgium
- VIB Center for Brain & Disease Research, Laboratory of Neurobiology, VIB-KU Leuven, Leuven, Belgium
| | - G Bultynck
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - A S Saab
- Institute of Pharmacology and Toxicology, Neuroscience Center Zurich, University of Zurich, University and ETH Zurich, Zurich, Switzerland
| | - E Wolfs
- Laboratory for Functional Imaging and Research on Stem Cells, BIOMED, UHasselt - Hasselt University, Diepenbeek, Belgium
| | - Y C Chai
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - U Himmelreich
- Biomedical MRI Group/MoSAIC, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - C Verfaillie
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - L Moons
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - L De Groef
- Cellular Communication and Neurodegeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium.
| |
Collapse
|
9
|
Nishida I, Yamada K, Sakamoto A, Wakabayashi T, Iwatsubo T. Chronic Neuronal Hyperexcitation Exacerbates Tau Propagation in a Mouse Model of Tauopathy. Int J Mol Sci 2024; 25:9004. [PMID: 39201689 PMCID: PMC11354494 DOI: 10.3390/ijms25169004] [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: 07/18/2024] [Revised: 08/10/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
The intracerebral spread of tau is a critical mechanism associated with functional decline in Alzheimer's disease (AD) and other tauopathies. Recently, a hypothesis has emerged suggesting that tau propagation is linked to functional neuronal connections, specifically driven by neuronal hyperactivity. However, experimental validation of this hypothesis remains limited. In this study, we investigated how tau propagation from the entorhinal cortex to the hippocampus, the neuronal circuit most susceptible to tau pathology in AD, is affected by the selective stimulation of neuronal activity along this circuit. Using a mouse model of seed-induced propagation combined with optogenetics, we found that the chronic stimulation of this neuronal connection over a 4-week period resulted in a significant increase in insoluble tau accumulation in both the entorhinal cortex and hippocampus. Importantly, the ratio of tau accumulation in the hippocampus relative to that in the entorhinal cortex, serving as an indicator of transcellular spreading, was significantly higher in mice subjected to chronic stimulation. These results support the notion that abnormal neuronal activity promotes tau propagation, thereby implicating it in the progression of tauopathy.
Collapse
Affiliation(s)
- Itaru Nishida
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo 1130033, Japan; (I.N.); (A.S.); (T.W.)
| | - Kaoru Yamada
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo 1130033, Japan; (I.N.); (A.S.); (T.W.)
| | - Asami Sakamoto
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo 1130033, Japan; (I.N.); (A.S.); (T.W.)
| | - Tomoko Wakabayashi
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo 1130033, Japan; (I.N.); (A.S.); (T.W.)
- Department of Pathophysiology, Meiji Pharmaceutical University, Tokyo 2040004, Japan
| | - Takeshi Iwatsubo
- National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 1878551, Japan;
| |
Collapse
|
10
|
Ondrejcak T, Klyubin I, Hu NW, Yang Y, Zhang Q, Rodriguez BJ, Rowan MJ. Rapidly reversible persistent long-term potentiation inhibition by patient-derived brain tau and amyloid ß proteins. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230234. [PMID: 38853565 PMCID: PMC11343230 DOI: 10.1098/rstb.2023.0234] [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: 10/20/2023] [Revised: 12/19/2023] [Accepted: 12/23/2023] [Indexed: 06/11/2024] Open
Abstract
How the two pathognomonic proteins of Alzheimer's disease (AD); amyloid ß (Aß) and tau, cause synaptic failure remains enigmatic. Certain synthetic and recombinant forms of these proteins are known to act concurrently to acutely inhibit long-term potentiation (LTP). Here, we examined the effect of early amyloidosis on the acute disruptive action of synaptotoxic tau prepared from recombinant protein and tau in patient-derived aqueous brain extracts. We also explored the persistence of the inhibition of LTP by different synaptotoxic tau preparations. A single intracerebral injection of aggregates of recombinant human tau that had been prepared by either sonication of fibrils (SτAs) or disulfide bond formation (oTau) rapidly and persistently inhibited LTP in rat hippocampus. The threshold for the acute inhibitory effect of oTau was lowered in amyloid precursor protein (APP)-transgenic rats. A single injection of synaptotoxic tau-containing AD or Pick's disease brain extracts also inhibited LTP, for over two weeks. Remarkably, the persistent disruption of synaptic plasticity by patient-derived brain tau was rapidly reversed by a single intracerebral injection of different anti-tau monoclonal antibodies, including one directed to a specific human tau amino acid sequence. We conclude that patient-derived LTP-disrupting tau species persist in the brain for weeks, maintaining their neuroactivity often in concert with Aß. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.
Collapse
Affiliation(s)
- Tomas Ondrejcak
- Department of Pharmacology and Therapeutics, School of Medicine, and Institute of Neuroscience, Trinity College, Dublin 2, Republic of Ireland
| | - Igor Klyubin
- Department of Pharmacology and Therapeutics, School of Medicine, and Institute of Neuroscience, Trinity College, Dublin 2, Republic of Ireland
| | - Neng-Wei Hu
- Department of Pharmacology and Therapeutics, School of Medicine, and Institute of Neuroscience, Trinity College, Dublin 2, Republic of Ireland
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou450001, People's Republic of China
| | - Yin Yang
- Department of Pharmacology and Therapeutics, School of Medicine, and Institute of Neuroscience, Trinity College, Dublin 2, Republic of Ireland
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou450001, People's Republic of China
| | - Qiancheng Zhang
- School of Physics and Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Republic of Ireland
| | - Brian J. Rodriguez
- School of Physics and Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Republic of Ireland
| | - Michael J. Rowan
- Department of Pharmacology and Therapeutics, School of Medicine, and Institute of Neuroscience, Trinity College, Dublin 2, Republic of Ireland
| |
Collapse
|
11
|
Sárkány B, Dávid C, Hortobágyi T, Gombás P, Somogyi P, Acsády L, Viney TJ. Early and selective localization of tau filaments to glutamatergic subcellular domains within the human anterodorsal thalamus. Acta Neuropathol 2024; 147:98. [PMID: 38861157 PMCID: PMC11166832 DOI: 10.1007/s00401-024-02749-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/21/2024] [Accepted: 06/01/2024] [Indexed: 06/12/2024]
Abstract
Widespread cortical accumulation of misfolded pathological tau proteins (ptau) in the form of paired helical filaments is a major hallmark of Alzheimer's disease. Subcellular localization of ptau at various stages of disease progression is likely to be informative of the cellular mechanisms involving its spread. Here, we found that the density of ptau within several distinct rostral thalamic nuclei in post-mortem human tissue (n = 25 cases) increased with the disease stage, with the anterodorsal nucleus (ADn) consistently being the most affected. In the ADn, ptau-positive elements were present already in the pre-cortical (Braak 0) stage. Tau pathology preferentially affected the calretinin-expressing subpopulation of glutamatergic neurons in the ADn. At the subcellular level, we detected ptau immunoreactivity in ADn cell bodies, dendrites, and in a specialized type of presynaptic terminal that expresses vesicular glutamate transporter 2 (vGLUT2) and likely originates from the mammillary body. The ptau-containing terminals displayed signs of degeneration, including endosomal/lysosomal organelles. In contrast, corticothalamic axon terminals lacked ptau. The data demonstrate the involvement of a specific cell population in ADn at the onset of the disease. The presence of ptau in subcortical glutamatergic presynaptic terminals supports hypotheses about the transsynaptic spread of tau selectively affecting specialized axonal pathways.
Collapse
Affiliation(s)
- Barbara Sárkány
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK.
| | - Csaba Dávid
- Lendület Laboratory of Thalamus Research, Institute of Experimental Medicine, Budapest, 1083, Hungary
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Tibor Hortobágyi
- Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Péter Gombás
- Department of Pathology, Szt. Borbála Hospital, Tatabánya, 2800, Hungary
| | - Peter Somogyi
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - László Acsády
- Lendület Laboratory of Thalamus Research, Institute of Experimental Medicine, Budapest, 1083, Hungary.
| | - Tim J Viney
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK.
| |
Collapse
|
12
|
Barbas H, Garcia-Cabezas MA, John Y, Bautista J, McKee A, Zikopoulos B. Cortical circuit principles predict patterns of trauma induced tauopathy in humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.02.592271. [PMID: 38746103 PMCID: PMC11092596 DOI: 10.1101/2024.05.02.592271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Connections in the cortex of diverse mammalian species are predicted reliably by the Structural Model for direction of pathways and signal processing (reviewed in 1,2). The model is rooted in the universal principle of cortical systematic variation in laminar structure and has been supported widely for connection patterns in animals but has not yet been tested for humans. Here, in postmortem brains of individuals neuropathologically diagnosed with chronic traumatic encephalopathy (CTE) we studied whether the hyperphosphorylated tau (p-tau) pathology parallels connection sequence in time by circuit mechanisms. CTE is a progressive p-tau pathology that begins focally in perivascular sites in sulcal depths of the neocortex (stages I-II) and later involves the medial temporal lobe (MTL) in stages III-IV. We provide novel quantitative evidence that the p-tau pathology in MTL A28 and nearby sites in CTE stage III closely follows the graded laminar patterns seen in homologous cortico-cortical connections in non-human primates. The Structural Model successfully predicted the laminar distribution of the p-tau neurofibrillary tangles and neurites and their density, based on the relative laminar (dis)similarity between the cortical origin (seed) and each connection site. The findings were validated for generalizability by a computational progression model. By contrast, the early focal perivascular pathology in the sulcal depths followed local columnar connectivity rules. These findings support the general applicability of a theoretical model to unravel the direction and progression of p-tau pathology in human neurodegeneration via a cortico-cortical mechanism. Cortical pathways converging on medial MTL help explain the progressive spread of p-tau pathology from focal cortical sites in early CTE to widespread lateral MTL areas and beyond in later disease stages.
Collapse
Affiliation(s)
- Helen Barbas
- Neural Systems Laboratory, Department of Health Sciences, Boston University, Boston, MA 022152
- Graduate Program in Neuroscience, Boston Univ. and School of Medicine
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA
- Center for Systems Neuroscience, Boston University, Boston, MA
| | - Miguel Angel Garcia-Cabezas
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Yohan John
- Neural Systems Laboratory, Department of Health Sciences, Boston University, Boston, MA 022152
| | - Julied Bautista
- Neural Systems Laboratory, Department of Health Sciences, Boston University, Boston, MA 022152
| | - Ann McKee
- Veterans Affairs (VA) Boston Healthcare System, US Department of Veteran Affairs, Boston, Massachusetts
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
| | - Basilis Zikopoulos
- Graduate Program in Neuroscience, Boston Univ. and School of Medicine
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA
- Center for Systems Neuroscience, Boston University, Boston, MA
- Human Systems Neuroscience Laboratory, Department of Health Sciences, Boston University
| |
Collapse
|
13
|
San Gil R, Pascovici D, Venturato J, Brown-Wright H, Mehta P, Madrid San Martin L, Wu J, Luan W, Chui YK, Bademosi AT, Swaminathan S, Naidoo S, Berning BA, Wright AL, Keating SS, Curtis MA, Faull RLM, Lee JD, Ngo ST, Lee A, Morsch M, Chung RS, Scotter E, Lisowski L, Mirzaei M, Walker AK. A transient protein folding response targets aggregation in the early phase of TDP-43-mediated neurodegeneration. Nat Commun 2024; 15:1508. [PMID: 38374041 PMCID: PMC10876645 DOI: 10.1038/s41467-024-45646-9] [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: 09/26/2023] [Accepted: 01/31/2024] [Indexed: 02/21/2024] Open
Abstract
Understanding the mechanisms that drive TDP-43 pathology is integral to combating amyotrophic lateral sclerosis (ALS), frontotemporal lobar degeneration (FTLD) and other neurodegenerative diseases. Here we generated a longitudinal quantitative proteomic map of the cortex from the cytoplasmic TDP-43 rNLS8 mouse model of ALS and FTLD, and developed a complementary open-access webtool, TDP-map ( https://shiny.rcc.uq.edu.au/TDP-map/ ). We identified distinct protein subsets enriched for diverse biological pathways with temporal alterations in protein abundance, including increases in protein folding factors prior to disease onset. This included increased levels of DnaJ homolog subfamily B member 5, DNAJB5, which also co-localized with TDP-43 pathology in diseased human motor cortex. DNAJB5 over-expression decreased TDP-43 aggregation in cell and cortical neuron cultures, and knockout of Dnajb5 exacerbated motor impairments caused by AAV-mediated cytoplasmic TDP-43 expression in mice. Together, these findings reveal molecular mechanisms at distinct stages of ALS and FTLD progression and suggest that protein folding factors could be protective in neurodegenerative diseases.
Collapse
Affiliation(s)
- Rebecca San Gil
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Dana Pascovici
- Insight Stats, Croydon Park, NSW, Australia
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde Sydney, NSW, Australia
| | - Juliana Venturato
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Heledd Brown-Wright
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Prachi Mehta
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- Motor Neuron Disease Research Centre, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Lidia Madrid San Martin
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Jemma Wu
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde Sydney, NSW, Australia
| | - Wei Luan
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Yi Kit Chui
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Adekunle T Bademosi
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Shilpa Swaminathan
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Serey Naidoo
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Britt A Berning
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Amanda L Wright
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Sean S Keating
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Maurice A Curtis
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - John D Lee
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Shyuan T Ngo
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Albert Lee
- Motor Neuron Disease Research Centre, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Marco Morsch
- Motor Neuron Disease Research Centre, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Roger S Chung
- Motor Neuron Disease Research Centre, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Emma Scotter
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Leszek Lisowski
- Vector and Genome Engineering Facility, Children's Medical Research Institute, Westmead, NSW, Australia
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine - National Research Institute, Warsaw, Poland
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Mehdi Mirzaei
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde Sydney, NSW, Australia
| | - Adam K Walker
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
14
|
Marei HE, Khan MUA, Hasan A. Potential use of iPSCs for disease modeling, drug screening, and cell-based therapy for Alzheimer's disease. Cell Mol Biol Lett 2023; 28:98. [PMID: 38031028 PMCID: PMC10687886 DOI: 10.1186/s11658-023-00504-2] [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: 08/26/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic illness marked by increasing cognitive decline and nervous system deterioration. At this time, there is no known medication that will stop the course of Alzheimer's disease; instead, most symptoms are treated. Clinical trial failure rates for new drugs remain high, highlighting the urgent need for improved AD modeling for improving understanding of the underlying pathophysiology of disease and improving drug development. The development of induced pluripotent stem cells (iPSCs) has made it possible to model neurological diseases like AD, giving access to an infinite number of patient-derived cells capable of differentiating neuronal fates. This advance will accelerate Alzheimer's disease research and provide an opportunity to create more accurate patient-specific models of Alzheimer's disease to support pathophysiological research, drug development, and the potential application of stem cell-based therapeutics. This review article provides a complete summary of research done to date on the potential use of iPSCs from AD patients for disease modeling, drug discovery, and cell-based therapeutics. Current technological developments in AD research including 3D modeling, genome editing, gene therapy for AD, and research on familial (FAD) and sporadic (SAD) forms of the disease are discussed. Finally, we outline the issues that need to be elucidated and future directions for iPSC modeling in AD.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35116, Egypt.
| | - Muhammad Umar Aslam Khan
- Biomedical Research Center, Qatar University, 2713, Doha, Qatar
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar
| |
Collapse
|
15
|
Haam J, Gunin S, Wilson L, Fry S, Bernstein B, Thomson E, Noblet H, Cushman J, Yakel JL. Entorhinal cortical delta oscillations drive memory consolidation. Cell Rep 2023; 42:113267. [PMID: 37838945 PMCID: PMC10872950 DOI: 10.1016/j.celrep.2023.113267] [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: 01/25/2023] [Revised: 08/30/2023] [Accepted: 09/28/2023] [Indexed: 10/17/2023] Open
Abstract
Long-term memories are formed by creating stable memory representations via memory consolidation, which mainly occurs during sleep following the encoding of labile memories in the hippocampus during waking. The entorhinal cortex (EC) has intricate connections with the hippocampus, but its role in memory consolidation is largely unknown. Using cell-type- and input-specific in vivo neural activity recordings, here we show that the temporoammonic pathway neurons in the EC, which directly innervate the output area of the hippocampus, exhibit potent oscillatory activities during anesthesia and sleep. Using in vivo individual and populational neuronal activity recordings, we demonstrate that a subpopulation of the temporoammonic pathway neurons, which we termed sleep cells, generate delta oscillations via hyperpolarization-activated cyclic-nucleotide-gated channels during sleep. The blockade of these oscillations significantly impaired the consolidation of hippocampus-dependent memory. Together, our findings uncover a key driver of delta oscillations and memory consolidation that are found in the EC.
Collapse
Affiliation(s)
- Juhee Haam
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA; Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC 27709, USA.
| | - Suman Gunin
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Leslie Wilson
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC 27709, USA
| | - Sydney Fry
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC 27709, USA
| | - Briana Bernstein
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC 27709, USA
| | - Eric Thomson
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC 27709, USA
| | - Hayden Noblet
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Jesse Cushman
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC 27709, USA
| | - Jerrel L Yakel
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
16
|
Grieco SF, Holmes TC, Xu X. Probing neural circuit mechanisms in Alzheimer's disease using novel technologies. Mol Psychiatry 2023; 28:4407-4420. [PMID: 36959497 PMCID: PMC10827671 DOI: 10.1038/s41380-023-02018-x] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/25/2023]
Abstract
The study of Alzheimer's Disease (AD) has traditionally focused on neuropathological mechanisms that has guided therapies that attenuate neuropathological features. A new direction is emerging in AD research that focuses on the progressive loss of cognitive function due to disrupted neural circuit mechanisms. Evidence from humans and animal models of AD show that dysregulated circuits initiate a cascade of pathological events that culminate in functional loss of learning, memory, and other aspects of cognition. Recent progress in single-cell, spatial, and circuit omics informs this circuit-focused approach by determining the identities, locations, and circuitry of the specific cells affected by AD. Recently developed neuroscience tools allow for precise access to cell type-specific circuitry so that their functional roles in AD-related cognitive deficits and disease progression can be tested. An integrated systems-level understanding of AD-associated neural circuit mechanisms requires new multimodal and multi-scale interrogations that longitudinally measure and/or manipulate the ensemble properties of specific molecularly-defined neuron populations first susceptible to AD. These newly developed technological and conceptual advances present new opportunities for studying and treating circuits vulnerable in AD and represent the beginning of a new era for circuit-based AD research.
Collapse
Affiliation(s)
- Steven F Grieco
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
- Center for Neural Circuit Mapping (CNCM), University of California, Irvine, CA, 92697, USA
| | - Todd C Holmes
- Center for Neural Circuit Mapping (CNCM), University of California, Irvine, CA, 92697, USA
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA.
- Center for Neural Circuit Mapping (CNCM), University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
17
|
Osanai H, Nair IR, Kitamura T. Dissecting cell-type-specific pathways in medial entorhinal cortical-hippocampal network for episodic memory. J Neurochem 2023; 166:172-188. [PMID: 37248771 PMCID: PMC10538947 DOI: 10.1111/jnc.15850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/07/2023] [Accepted: 05/10/2023] [Indexed: 05/31/2023]
Abstract
Episodic memory, which refers to our ability to encode and recall past events, is essential to our daily lives. Previous research has established that both the entorhinal cortex (EC) and hippocampus (HPC) play a crucial role in the formation and retrieval of episodic memories. However, to understand neural circuit mechanisms behind these processes, it has become necessary to monitor and manipulate the neural activity in a cell-type-specific manner with high temporal precision during memory formation, consolidation, and retrieval in the EC-HPC networks. Recent studies using cell-type-specific labeling, monitoring, and manipulation have demonstrated that medial EC (MEC) contains multiple excitatory neurons that have differential molecular markers, physiological properties, and anatomical features. In this review, we will comprehensively examine the complementary roles of superficial layers of neurons (II and III) and the roles of deeper layers (V and VI) in episodic memory formation and recall based on these recent findings.
Collapse
Affiliation(s)
- Hisayuki Osanai
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Indrajith R Nair
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Takashi Kitamura
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
18
|
Chen S, Acosta D, Fu H. New unexpected role for Wolfram Syndrome protein WFS1: a novel therapeutic target for Alzheimer's disease? Neural Regen Res 2023; 18:1501-1502. [PMID: 36571353 PMCID: PMC10075124 DOI: 10.4103/1673-5374.361540] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/20/2022] [Accepted: 10/29/2022] [Indexed: 11/19/2022] Open
Affiliation(s)
- Shuo Chen
- Department of Neuroscience, Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Diana Acosta
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Hongjun Fu
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
19
|
Qu W, Canoll P, Hargus G. Molecular Insights into Cell Type-specific Roles in Alzheimer's Disease: Human Induced Pluripotent Stem Cell-based Disease Modelling. Neuroscience 2023; 518:10-26. [PMID: 35569647 PMCID: PMC9974106 DOI: 10.1016/j.neuroscience.2022.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 10/18/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia resulting in widespread degeneration of the central nervous system with severe cognitive impairment. Despite the devastating toll of AD, the incomplete understanding of the complex molecular mechanisms hinders the expeditious development of effective cures. Emerging evidence from animal studies has shown that different brain cell types play distinct roles in the pathogenesis of AD. Glutamatergic neurons are preferentially affected in AD and pronounced gliosis contributes to the progression of AD in both a cell-autonomous and a non-cell-autonomous manner. Much has been discovered through genetically modified animal models, yet frequently failed translational attempts to clinical applications call for better disease models. Emerging evidence supports the significance of human-induced pluripotent stem cell (iPSC) derived brain cells in modeling disease development and progression, opening new avenues for the discovery of molecular mechanisms. This review summarizes the function of different cell types in the pathogenesis of AD, such as neurons, microglia, and astrocytes, and recognizes the potential of utilizing the rapidly growing iPSC technology in modeling AD.
Collapse
Affiliation(s)
- Wenhui Qu
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, United States
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Gunnar Hargus
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, United States.
| |
Collapse
|
20
|
Tang Y, Yan Y, Mao J, Ni J, Qing H. The hippocampus associated GABAergic neural network impairment in early-stage of Alzheimer's disease. Ageing Res Rev 2023; 86:101865. [PMID: 36716975 DOI: 10.1016/j.arr.2023.101865] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/13/2023] [Accepted: 01/25/2023] [Indexed: 01/29/2023]
Abstract
Alzheimer's disease (AD) is the commonest neurodegenerative disease with slow progression. Pieces of evidence suggest that the GABAergic system is impaired in the early stage of AD, leading to hippocampal neuron over-activity and further leading to memory and cognitive impairment in patients with AD. However, the precise impairment mechanism of the GABAergic system on the pathogenesis of AD is still unclear. The impairment of neural networks associated with the GABAergic system is tightly associated with AD. Therefore, we describe the roles played by hippocampus-related GABAergic circuits and their impairments in AD neuropathology. In addition, we give our understand on the process from GABAergic circuit impairment to cognitive and memory impairment, since recent studies on astrocyte in AD plays an important role behind cognition dysfunction caused by GABAergic circuit impairment, which helps better understand the GABAergic system and could open up innovative AD therapy.
Collapse
Affiliation(s)
- Yuanhong Tang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Yan Yan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Jian Mao
- Zhengzhou Tobacco Institute of China National Tobacco Company, Zhengzhou 450001, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; Department of Biology, Shenzhen MSU-BIT University, Shenzhen 518172, China.
| |
Collapse
|
21
|
Punapart M, Reimets R, Seppa K, Kirillov S, Gaur N, Eskla KL, Jagomäe T, Vasar E, Plaas M. Chronic Stress Alters Hippocampal Renin-Angiotensin-Aldosterone System Component Expression in an Aged Rat Model of Wolfram Syndrome. Genes (Basel) 2023; 14:genes14040827. [PMID: 37107585 PMCID: PMC10137641 DOI: 10.3390/genes14040827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 04/01/2023] Open
Abstract
Biallelic mutations in the gene encoding WFS1 underlie the development of Wolfram syndrome (WS), a rare neurodegenerative disorder with no available cure. We have previously shown that Wfs1 deficiency can impair the functioning of the renin-angiotensin-aldosterone system (RAAS). The expression of two key receptors, angiotensin II receptor type 2 (Agtr2) and bradykinin receptor B1 (Bdkrb1), was downregulated both in vitro and in vivo across multiple organs in a rat model of WS. Here, we show that the expression of key RAAS components is also dysregulated in neural tissue from aged WS rats and that these alterations are not normalized by pharmacological treatments (liraglutide (LIR), 7,8-dihydroxyflavone (7,8-DHF) or their combination). We found that the expression of angiotensin II receptor type 1a (Agtr1a), angiotensin II receptor type 1b (Agtr1b), Agtr2 and Bdkrb1 was significantly downregulated in the hippocampus of WS animals that experienced chronic experimental stress. Treatment-naïve WS rats displayed different gene expression patterns, underscoring the effect of prolonged experiment-induced stress. Altogether, we posit that Wfs1 deficiency disturbs RAAS functioning under chronic stressful conditions, thereby exacerbating neurodegeneration in WS.
Collapse
Affiliation(s)
- Marite Punapart
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411 Tartu, Estonia
| | - Riin Reimets
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411 Tartu, Estonia
| | - Kadri Seppa
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411 Tartu, Estonia
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
| | - Silvia Kirillov
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411 Tartu, Estonia
| | - Nayana Gaur
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411 Tartu, Estonia
| | - Kattri-Liis Eskla
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
| | - Toomas Jagomäe
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411 Tartu, Estonia
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
| | - Mario Plaas
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411 Tartu, Estonia
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
- Correspondence:
| |
Collapse
|
22
|
Chen Y, Zhang M, Zhou Y, Li P. Case Report: A novel mutation in WFS1 gene (c.1756G>A p.A586T) is responsible for early clinical features of cognitive impairment and recurrent ischemic stroke. Front Genet 2023; 14:1072978. [PMID: 36816038 PMCID: PMC9932685 DOI: 10.3389/fgene.2023.1072978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/06/2023] [Indexed: 02/05/2023] Open
Abstract
Wolfram syndrome 1 (WFS1) gene mutations can be dominantly or recessively inherited, and the onset of the clinical picture is highly heterogeneity in both appearance and degree of severity. Different types of WFS1 mutations have been identified. Autosomal recessive mutations in the WFS1 gene will underlie Wolfram syndrome 1 (WS1), a rare and severe neurodegenerative disease characterized by diabetes insipidus, diabetes mellitus, optic atrophy, deafness, and other neurological, urological and psychiatric abnormalities. Other WFS1-related disorders such as low-frequency sensorineural hearing impairment (LFSNHI) and Wolfram syndrome-like disease with autosomal dominant transmission have been described. It is difficult to establish genotype-phenotype correlations because of the molecular complexity of wolframin protein. In this report, we presented a case of WSF1 gene mutation-related disease with cognitive impairment as the initial symptom and recurrent cerebral infarction in the course of the disease. Brain structural imaging results suggested decreased intracranial volume, dramatically reduced in cerebral cortex and cerebellum regions. Multimodal molecular imaging results suggested Tau protein deposition in the corresponding brain regions without Aβ pathology changes. These pathological changes may indicate a role of WFS1 in neuronal vulnerability to tau pathology associated with neurodegeneration and ischemia-induced damage.
Collapse
Affiliation(s)
- Yuan Chen
- Department of Neurology, Tianjin Huanhu Hospital, Clinical College of Neurology, Neurosurgery, and Neurorehabilitation, Tianjin Medical University, Tianjin, China,Department of Neurology, Tianjin Huanhu Hospital affiliated to Nankai University, Tianjin University Huanhu Hospital, Tianjin, China,Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgery Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Miao Zhang
- Department of Neurology, Tianjin Huanhu Hospital, Clinical College of Neurology, Neurosurgery, and Neurorehabilitation, Tianjin Medical University, Tianjin, China,Department of Neurology, Tianjin Huanhu Hospital affiliated to Nankai University, Tianjin University Huanhu Hospital, Tianjin, China,Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgery Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Yuying Zhou
- Department of Neurology, Tianjin Huanhu Hospital, Clinical College of Neurology, Neurosurgery, and Neurorehabilitation, Tianjin Medical University, Tianjin, China,Department of Neurology, Tianjin Huanhu Hospital affiliated to Nankai University, Tianjin University Huanhu Hospital, Tianjin, China,Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgery Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Pan Li
- Department of Neurology, Tianjin Huanhu Hospital, Clinical College of Neurology, Neurosurgery, and Neurorehabilitation, Tianjin Medical University, Tianjin, China,Department of Neurology, Tianjin Huanhu Hospital affiliated to Nankai University, Tianjin University Huanhu Hospital, Tianjin, China,Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgery Institute, Tianjin Huanhu Hospital, Tianjin, China,*Correspondence: Pan Li,
| |
Collapse
|
23
|
Rossi G, Ordazzo G, Vanni NN, Castoldi V, Iannielli A, Di Silvestre D, Bellini E, Bernardo L, Giannelli SG, Luoni M, Muggeo S, Leocani L, Mauri P, Broccoli V. MCT1-dependent energetic failure and neuroinflammation underlie optic nerve degeneration in Wolfram syndrome mice. eLife 2023; 12:81779. [PMID: 36645345 PMCID: PMC9891717 DOI: 10.7554/elife.81779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/13/2023] [Indexed: 01/17/2023] Open
Abstract
Wolfram syndrome 1 (WS1) is a rare genetic disorder caused by mutations in the WFS1 gene leading to a wide spectrum of clinical dysfunctions, among which blindness, diabetes, and neurological deficits are the most prominent. WFS1 encodes for the endoplasmic reticulum (ER) resident transmembrane protein wolframin with multiple functions in ER processes. However, the WFS1-dependent etiopathology in retinal cells is unknown. Herein, we showed that Wfs1 mutant mice developed early retinal electrophysiological impairments followed by marked visual loss. Interestingly, axons and myelin disruption in the optic nerve preceded the degeneration of the retinal ganglion cell bodies in the retina. Transcriptomics at pre-degenerative stage revealed the STAT3-dependent activation of proinflammatory glial markers with reduction of the homeostatic and pro-survival factors glutamine synthetase and BDNF. Furthermore, label-free comparative proteomics identified a significant reduction of the monocarboxylate transport isoform 1 (MCT1) and its partner basigin that are highly enriched on retinal glia and myelin-forming oligodendrocytes in optic nerve together with wolframin. Loss of MCT1 caused a failure in lactate transfer from glial to neuronal cell bodies and axons leading to a chronic hypometabolic state. Thus, this bioenergetic impairment is occurring concurrently both within the axonal regions and cell bodies of the retinal ganglion cells, selectively endangering their survival while impacting less on other retinal cells. This metabolic dysfunction occurs months before the frank RGC degeneration suggesting an extended time-window for intervening with new therapeutic strategies focused on boosting retinal and optic nerve bioenergetics in WS1.
Collapse
Affiliation(s)
- Greta Rossi
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
| | - Gabriele Ordazzo
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
| | - Niccolò N Vanni
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
| | - Valerio Castoldi
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
- Experimental Neurophysiology Unit, Institute of Experimental Neurology (INSPE), San Raffaele Scientific InstituteMilanItaly
| | - Angelo Iannielli
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
- National Research Council of Italy, Institute of NeuroscienceMilanoItaly
| | - Dario Di Silvestre
- National Research Council of Italy, Institute of Technologies in BiomedicineMilanItaly
| | - Edoardo Bellini
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
| | - Letizia Bernardo
- National Research Council of Italy, Institute of Technologies in BiomedicineMilanItaly
| | | | - Mirko Luoni
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
- National Research Council of Italy, Institute of NeuroscienceMilanoItaly
| | - Sharon Muggeo
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
| | - Letizia Leocani
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
- Experimental Neurophysiology Unit, Institute of Experimental Neurology (INSPE), San Raffaele Scientific InstituteMilanItaly
| | - PierLuigi Mauri
- National Research Council of Italy, Institute of Technologies in BiomedicineMilanItaly
| | - Vania Broccoli
- Division of Neuroscience, San Raffaele Scientific InstituteMilanoItaly
- National Research Council of Italy, Institute of NeuroscienceMilanoItaly
| |
Collapse
|
24
|
Emerging Roles of Extracellular Vesicles in Alzheimer's Disease: Focus on Synaptic Dysfunction and Vesicle-Neuron Interaction. Cells 2022; 12:cells12010063. [PMID: 36611856 PMCID: PMC9818402 DOI: 10.3390/cells12010063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Alzheimer's disease (AD) is considered by many to be a synaptic failure. Synaptic function is in fact deeply affected in the very early disease phases and recognized as the main cause of AD-related cognitive impairment. While the reciprocal involvement of amyloid beta (Aβ) and tau peptides in these processes is under intense investigation, the crucial role of extracellular vesicles (EVs) released by different brain cells as vehicles for these molecules and as mediators of early synaptic alterations is gaining more and more ground in the field. In this review, we will summarize the current literature on the contribution of EVs derived from distinct brain cells to neuronal alterations and build a working model for EV-mediated propagation of synaptic dysfunction in early AD. A deeper understanding of EV-neuron interaction will provide useful targets for the development of novel therapeutic approaches aimed at hampering AD progression.
Collapse
|
25
|
Wang Z, Wang X, Shi L, Cai Y, Hu B. Wolfram syndrome 1b mutation suppresses Mauthner-cell axon regeneration via ER stress signal pathway. Acta Neuropathol Commun 2022; 10:184. [PMID: 36527091 PMCID: PMC9758940 DOI: 10.1186/s40478-022-01484-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
Abstract
Wolfram Syndrome (WS) is a fatal human inherited disease with symptoms of diabetes, vision decreasing, and neurodegeneration caused by mutations in the endoplasmic reticulum (ER)-resident protein WFS1. WFS1 has been reported to play an important role in glucose metabolism. However, the role of WFS1 in axonal regeneration in the central nervous system has so far remained elusive. Herein, we established a model of the wfs1b globally deficient zebrafish line. wfs1b deficiency severely impeded the Mauthner-cell (M-cell) axon regeneration, which was partly dependent on the ER stress response. The administration of ER stress inhibitor 4-Phenylbutyric acid (4-PBA) promoted M-cell axon regeneration in wfs1b-/- zebrafish larvae, while the ER stress activator Tunicamycin (TM) inhibited M-cell axon regeneration in wfs1b+/+ zebrafish larvae. Moreover, complementation of wfs1b at the single-cell level stimulated M-cell axon regeneration in the wfs1b-/- zebrafish larvae. Altogether, our results revealed that wfs1b promotes M-cell axon regeneration through the ER stress signal pathway and provide new evidence for a therapeutic target for WS and axon degeneration.
Collapse
Affiliation(s)
- Zongyi Wang
- grid.59053.3a0000000121679639Hefei National Research Center for Physical Sciences at the Microscale, Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 China
| | - Xinliang Wang
- grid.59053.3a0000000121679639Hefei National Research Center for Physical Sciences at the Microscale, Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 China
| | - Lingyu Shi
- grid.59053.3a0000000121679639Hefei National Research Center for Physical Sciences at the Microscale, Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 China
| | - Yuan Cai
- grid.59053.3a0000000121679639Hefei National Research Center for Physical Sciences at the Microscale, Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 China ,grid.59053.3a0000000121679639First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 China
| | - Bing Hu
- grid.59053.3a0000000121679639Hefei National Research Center for Physical Sciences at the Microscale, Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 China ,grid.59053.3a0000000121679639Research Institute of Frontier Cross Science and Biomedical Sciences, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 China
| |
Collapse
|
26
|
Zhao R, Grunke SD, Wood CA, Perez GA, Comstock M, Li MH, Singh AK, Park KW, Jankowsky JL. Activity disruption causes degeneration of entorhinal neurons in a mouse model of Alzheimer's circuit dysfunction. eLife 2022; 11:e83813. [PMID: 36468693 PMCID: PMC9873254 DOI: 10.7554/elife.83813] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases are characterized by selective vulnerability of distinct cell populations; however, the cause for this specificity remains elusive. Here, we show that entorhinal cortex layer 2 (EC2) neurons are unusually vulnerable to prolonged neuronal inactivity compared with neighboring regions of the temporal lobe, and that reelin + stellate cells connecting EC with the hippocampus are preferentially susceptible within the EC2 population. We demonstrate that neuronal death after silencing can be elicited through multiple independent means of activity inhibition, and that preventing synaptic release, either alone or in combination with electrical shunting, is sufficient to elicit silencing-induced degeneration. Finally, we discovered that degeneration following synaptic silencing is governed by competition between active and inactive cells, which is a circuit refinement process traditionally thought to end early in postnatal life. Our data suggests that the developmental window for wholesale circuit plasticity may extend into adulthood for specific brain regions. We speculate that this sustained potential for remodeling by entorhinal neurons may support lifelong memory but renders them vulnerable to prolonged activity changes in disease.
Collapse
Affiliation(s)
- Rong Zhao
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Stacy D Grunke
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Caleb A Wood
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Gabriella A Perez
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Melissa Comstock
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Ming-Hua Li
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Anand K Singh
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Kyung-Won Park
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Joanna L Jankowsky
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
- Departments of Neurology, Neurosurgery, and Molecular and Cellular Biology, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
27
|
Chen S, Acosta D, Li L, Liang J, Chang Y, Wang C, Fitzgerald J, Morrison C, Goulbourne CN, Nakano Y, Villegas NCH, Venkataraman L, Brown C, Serrano GE, Bell E, Wemlinger T, Wu M, Kokiko-Cochran ON, Popovich P, Flowers XE, Honig LS, Vonsattel JP, Scharre DW, Beach TG, Ma Q, Kuret J, Kõks S, Urano F, Duff KE, Fu H. Wolframin is a novel regulator of tau pathology and neurodegeneration. Acta Neuropathol 2022; 143:547-569. [PMID: 35389045 DOI: 10.1007/s00401-022-02417-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 12/17/2022]
Abstract
Selective neuronal vulnerability to protein aggregation is found in many neurodegenerative diseases including Alzheimer's disease (AD). Understanding the molecular origins of this selective vulnerability is, therefore, of fundamental importance. Tau protein aggregates have been found in Wolframin (WFS1)-expressing excitatory neurons in the entorhinal cortex, one of the earliest affected regions in AD. The role of WFS1 in Tauopathies and its levels in tau pathology-associated neurodegeneration, however, is largely unknown. Here we report that WFS1 deficiency is associated with increased tau pathology and neurodegeneration, whereas overexpression of WFS1 reduces those changes. We also find that WFS1 interacts with tau protein and controls the susceptibility to tau pathology. Furthermore, chronic ER stress and autophagy-lysosome pathway (ALP)-associated genes are enriched in WFS1-high excitatory neurons in human AD at early Braak stages. The protein levels of ER stress and autophagy-lysosome pathway (ALP)-associated proteins are changed in tau transgenic mice with WFS1 deficiency, while overexpression of WFS1 reverses those changes. This work demonstrates a possible role for WFS1 in the regulation of tau pathology and neurodegeneration via chronic ER stress and the downstream ALP. Our findings provide insights into mechanisms that underpin selective neuronal vulnerability, and for developing new therapeutics to protect vulnerable neurons in AD.
Collapse
|