1
|
Alves RL, Gonçalves A, Voytyuk I, Harrison DC. Behaviour profile characterization of PS19 and rTg4510 tauopathy mouse models: A systematic review and a meta-analysis. Exp Neurol 2025; 389:115234. [PMID: 40185359 DOI: 10.1016/j.expneurol.2025.115234] [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: 01/17/2025] [Revised: 03/14/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
The rTg4510 and PS19 mouse models are widely used in tauopathy research. Alzheimer's disease (AD) is the most prevalent among tauopathies. Behavioural tests are frequently used to assess emotional, cognitive, and motor behaviours in mouse models of AD. Cognitive deficits begin to manifest in rTg4510 mice around 3 months of age and in PS19 mice around 6 months. However, it's widely recognized that behavioural outcomes can vary due to environmental factors, health status, and husbandry practices, causing phenotypic differences between facilities. This study aims to consolidate current knowledge of the behavioural phenotypes of these two mouse models. We conducted a comprehensive literature review using keyword searches with Boolean operators across databases up to January 2024. Additional studies were included from manual searches. A total of 23 articles were reviewed for rTg4510 mice and 52 for PS19 mice. We extracted methodological details and key findings from each study. Results for rTg4510 mice show consistent findings regarding locomotion, memory and learning, and neurological dysfunction. However, the limited studies on motor and balance behaviour revealed no significant differences, while anxiety-like behaviour showed some inconsistencies. PS19 mice demonstrate more robust results for anxiety-like behaviour, memory and learning, and locomotion, while findings for balance and coordination are more inconsistent. Although there is overall coherence in certain aspects of the behavioural profiles of these tauopathy mouse models, it is crucial to recognize experimental heterogeneity and profile behavioural baselines to optimize the testing of both genetic and pharmacological interventions.
Collapse
Affiliation(s)
- Renata L Alves
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, United Kingdom.
| | | | - Iryna Voytyuk
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, United Kingdom
| | - David C Harrison
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, United Kingdom
| |
Collapse
|
2
|
Engel S, Dillerud M, Scalf M, Dobbelmann R, Du Y, Lee AM, Graves SM. Alcohol consumption during early adulthood increases the vulnerability of locus coeruleus neurons and amyloid beta pathology in female APP/PS1 mice. Alcohol 2025:S0741-8329(25)00068-0. [PMID: 40409612 DOI: 10.1016/j.alcohol.2025.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 05/14/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025]
Abstract
Alcohol use disorder is the most common substance misuse disorder and Alzheimer's disease (AD) is the most common neurodegenerative disease. Evidence suggests that alcohol consumption may increase the risk for developing dementia and AD. The locus coeruleus (LC) is a region wherein the impact of alcohol and AD may converge. The LC is a noradrenergic nucleus that is highly vulnerable to degeneration in AD, and loss of LC neurons is associated with increased amyloid beta (Abeta) pathology. The present study examined whether alcohol consumption during early adulthood impacts LC degeneration and Abeta using the APP/PS1 mouse model. Female APP/PS1 mice underwent an eight-week chronic intermittent access (IA) alcohol consumption paradigm followed by twenty-three weeks of abstinence; water-consuming control subjects were run in parallel. APP/PS1 mice that had IA to alcohol showed a 21.9% decrease in the number of LC neurons and a decrease in the length of noradrenergic axons innervating the primary motor cortex. Furthermore, this alcohol induced LC deficit was associated with an increase in Abeta pathology in the primary motor cortex. In contrast to results from female APP/PS1 mice, there were no deficits in axon length and only a 9.4% decrease in the number of LC neurons in non-transgenic female subjects after abstinence from IA to alcohol. Our results demonstrate that alcohol consumption during early adulthood increases the vulnerability of LC neurons to degeneration and exacerbates Abeta pathology in female APP/PS1 mice, providing evidence that a history of alcohol abuse may impact the trajectory and severity of AD.
Collapse
Affiliation(s)
- Shaydel Engel
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455
| | - Madison Dillerud
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455
| | - Matthew Scalf
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455
| | - Ruth Dobbelmann
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455
| | - Yijuan Du
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455
| | - Anna M Lee
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455
| | - Steven M Graves
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455.
| |
Collapse
|
3
|
Nong Y, Kim JS, Jia L, Arancio O, Wang Q. The interaction between neurotransmitter receptor activity and amyloid-β pathology in Alzheimer's disease. J Alzheimers Dis 2025:13872877251342273. [PMID: 40388923 DOI: 10.1177/13872877251342273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
The accumulation of amyloid-β (Aβ) peptides is a hallmark of Alzheimer's disease (AD). Central to AD pathology is the production of Aβ peptides through the amyloidogenic processing of amyloid-β protein precursor (AβPP) by β-secretase (BACE-1) and γ-secretase. Recent studies have shifted focus from Aβ plaque deposits to the more toxic soluble Aβ oligomers. One significant way in which Aβ peptides impair neuronal information processing is by influencing neurotransmitter receptor function. These receptors, including adrenergic, acetylcholine, dopamine, 5-HT, glutamate, and gamma-aminobutyric acid (GABA) receptors, play a crucial role in regulating synaptic transmission, which underlies perceptual and cognitive functions. This review explores how Aβ interacts with these key neurotransmitter receptors and how these interactions contribute to neural dysfunction in AD. Moreover, we examine how agonists and antagonists of these receptors influence Aβ pathology, offering new perspectives on potential therapeutic strategies to curb AD progression effectively and improve patients' quality of life.
Collapse
Affiliation(s)
- Yuhan Nong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Jung Soo Kim
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Litian Jia
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Ottavio Arancio
- Departments of Pathology & Cell Biology, and Medicine, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Qi Wang
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Neurosurgery, Columbia University, New York, NY, USA
| |
Collapse
|
4
|
Downs AM, Kmiec G, Catavero CM, Wykoff LA, McElligott ZA. Loss of excitatory inputs and decreased tonic and evoked activity of locus coeruleus neurons in aged P301S mice. Neurobiol Dis 2025; 208:106883. [PMID: 40122182 PMCID: PMC12056759 DOI: 10.1016/j.nbd.2025.106883] [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: 01/17/2025] [Revised: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 03/25/2025] Open
Abstract
Tau pathology in the locus coeruleus (LC) is associated with several neurodegenerative conditions including Alzheimer's disease and frontotemporal dementia. Phosphorylated tau accumulates in the LC and results in inflammation, synaptic loss, and eventually cell death as the disease progresses. Loss of LC neurons and noradrenergic innervation is thought to contribute to the symptoms of cognitive decline later in disease. While loss and degeneration of LC neurons has been well studied, less is known about changes in LC physiology at advanced stages of tau pathology that precedes neurodegeneration. In this study, we investigated the ex vivo electrophysiological properties of LC neurons in male and female mice from the P301S mouse model of tauopathy at 9 months of age, a time-point when significant tau accumulation, cell death, and cognitive impairments are observed. We found a reduction in excitatory inputs and changes in excitatory post-synaptic current kinetics in male and female P301S. There was also a decrease in spontaneous discharge of LC neurons and an increase in AP threshold in P301S mice of both sexes. Finally, we observed a decrease in excitability and increase in rheobase current in P301S mice. Despite the decrease in LC activity in ex vivo slices, we did not identify differences in total tissue norepinephrine (NE) or NE metabolites in prefrontal cortex or hippocampus. Together these findings demonstrate reductions in the activity and excitability of LC neurons at late stages of tau accumulation. However, compensatory mechanisms may maintain normal NE levels in LC projection regions in vivo.
Collapse
Affiliation(s)
- Anthony M Downs
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States of America; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States of America.
| | - Gracianne Kmiec
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States of America
| | - Christina M Catavero
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States of America
| | - Luke A Wykoff
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States of America
| | - Zoé A McElligott
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States of America; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States of America; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States of America.
| |
Collapse
|
5
|
Wu J, Toporek A, Lin Q, Goldstein FC, Loring DW, Kelberman MA, Weinshenker D, Levey AI, Lah JJ, Qiu D. Probing locus coeruleus functional network in healthy aging and its association with Alzheimer's disease biomarkers using pupillometry. Alzheimers Res Ther 2025; 17:53. [PMID: 40016783 PMCID: PMC11866666 DOI: 10.1186/s13195-025-01701-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/18/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is the leading cause of dementia, and the early detection of the disease-associated changes allows early interventions. The locus coeruleus (LC) has been reported to be the first brain region to develop tau pathology in AD. However, the functional brain network of the LC in both healthy aging and AD pathology is largely unknown due to technical difficulties associated with the small size of the LC. In this study, we used the measurement of spontaneous pupil constriction/dilation as a surrogate for LC activity to study LC brain network changes during healthy aging. METHODS Thirty-seven healthy younger and thirty-nine healthy older adults were included from the Emory Healthy Brain Study and underwent resting-state functional MRI while simultaneously tracking pupil diameter. The measurements of pupil diameter dynamics were used as reference signals in brain connectivity analysis. The connectivity of the identified networks was then compared between younger and older participants. Correlations of the identified regions with neuropsychological assessments and cerebrospinal fluid (CSF) biomarkers were also evaluated. RESULTS A brain network of 20 clusters associated with pupil diameter dynamics was identified, including the LC as well as brain regions functionally connected to the LC. The pupil diameter network was found to positively correlate with the salience network and negatively correlate with the central executive network. Functional connectivity decreased within the pupil diameter network with healthy aging. The pupil diameter connectivity was associated with memory, executive, and visuospatial functioning. CSF total tau closely correlated with pupil diameter network. CONCLUSIONS Pupil diameter dynamics provide valuable insights into LC-related processes. While they are not solely influenced by LC activity, spontaneous pupil constrictor/dilatory activity shows promise as a non-invasive approach to probe the LC network and warrants further studies to evaluate its value as an early biomarker of AD.
Collapse
Affiliation(s)
- Junjie Wu
- Department of Radiology and Imaging Sciences, School of Medicine, Emory University, 1364 Clifton Rd NE, Atlanta, GA, 30322, USA
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Aaron Toporek
- Department of Radiology and Imaging Sciences, School of Medicine, Emory University, 1364 Clifton Rd NE, Atlanta, GA, 30322, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Qixiang Lin
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Felicia C Goldstein
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA
- Goizueta Alzheimer's Disease Research Center, Emory University, Atlanta, GA, USA
| | - David W Loring
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA
- Goizueta Alzheimer's Disease Research Center, Emory University, Atlanta, GA, USA
| | - Michael A Kelberman
- Department of Human Genetics, School of Medicine, Emory University, Atlanta, GA, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - David Weinshenker
- Goizueta Alzheimer's Disease Research Center, Emory University, Atlanta, GA, USA
- Department of Human Genetics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Allan I Levey
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA
- Goizueta Alzheimer's Disease Research Center, Emory University, Atlanta, GA, USA
| | - James J Lah
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA
- Goizueta Alzheimer's Disease Research Center, Emory University, Atlanta, GA, USA
| | - Deqiang Qiu
- Department of Radiology and Imaging Sciences, School of Medicine, Emory University, 1364 Clifton Rd NE, Atlanta, GA, 30322, USA.
- Goizueta Alzheimer's Disease Research Center, Emory University, Atlanta, GA, USA.
- Joint Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
6
|
Crawford JL, Berry AS. Examining resilience to Alzheimer's disease through the lens of monoaminergic neuromodulator systems. Trends Neurosci 2024; 47:892-903. [PMID: 39368845 PMCID: PMC11563896 DOI: 10.1016/j.tins.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/15/2024] [Accepted: 09/16/2024] [Indexed: 10/07/2024]
Abstract
The monoaminergic nuclei are thought to be some of the earliest sites of Alzheimer's disease (AD) pathology in the brain, with tau-containing pretangles appearing in these nuclei decades before the onset of clinical impairments. It has increasingly been recognized that monoamine systems represent a critical target of investigation towards understanding the progression of AD and designing early detection and treatment approaches. This review synthesizes evidence across animal studies, human neuropathology, and state-of-the-art neuroimaging and daily life assessment methods in humans, which demonstrate robust relationships between monoamine systems and AD pathophysiology and behavior. Further, the review highlights the promise of multimethod, multisystem approaches to studying monoaminergic mechanisms of resilience to AD pathology.
Collapse
Affiliation(s)
| | - Anne S Berry
- Department of Psychology, Brandeis University, Waltham, MA, USA.
| |
Collapse
|
7
|
Kelberman MA, Rodberg E, Arabzadeh E, Bair-Marshall CJ, Berridge CW, Berrocoso E, Breton-Provencher V, Chandler DJ, Che A, Davy O, Devilbiss DM, Downs AM, Drummond G, Dvorkin R, Fazlali Z, Froemke RC, Glennon E, Gold JI, Ito H, Jiang X, Johansen JP, Kaye AP, Kim JR, Kuo CC, Liu RJ, Liu Y, Llorca-Torralba M, McCall JG, McElligott ZA, McKinney AM, Miguelez C, Min MY, Nowlan AC, Omrani M, Poe GR, Pickering AE, Ranjbar-Slamloo Y, Razquin J, Rodenkirch C, Sales AC, Satyasambit R, Shea SD, Sur M, Tkaczynski JA, Torres-Sanchez S, Uematsu A, Vazquez CR, Vreven A, Wang Q, Waterhouse BD, Yang HW, Yang JH, Zhao L, Zouridis IS, Weinshenker D, Vazey E, Totah NK. Diversity of ancestral brainstem noradrenergic neurons across species and multiple biological factors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618224. [PMID: 39464004 PMCID: PMC11507722 DOI: 10.1101/2024.10.14.618224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The brainstem region, locus coeruleus (LC), has been remarkably conserved across vertebrates. Evolution has woven the LC into wide-ranging neural circuits that influence functions as broad as autonomic systems, the stress response, nociception, sleep, and high-level cognition among others. Given this conservation, there is a strong possibility that LC activity is inherently similar across species, and furthermore that age, sex, and brain state influence LC activity similarly across species. The degree to which LC activity is homogenous across these factors, however, has never been assessed due to the small sample size of individual studies. Here, we pool data from 20 laboratories (1,855 neurons) and show diversity across both intrinsic and extrinsic factors such as species, age, sex and brain state. We use a negative binomial regression model to compare activity from male monkeys, and rats and mice of both sexes that were recorded across brain states from brain slices ex vivo or under different anesthetics or during wakefulness in vivo. LC activity differed due to complex interactions of species, sex, and brain state. The LC became more active during aging, independent of sex. Finally, in contrast to the foundational principle that all species express two distinct LC firing modes ("tonic" or "phasic"), we discovered great diversity within spontaneous LC firing patterns. Different factors were associated with higher incidence of some firing modes. We conclude that the activity of the evolutionarily-ancient LC is not conserved. Inherent differences due to age and species-sex-brain state interactions have implications for understanding the role of LC in species-specific naturalistic behavior, as well as in psychiatric disorders, cardiovascular disease, immunology, and metabolic disorders.
Collapse
Affiliation(s)
- Michael A. Kelberman
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | - Ellen Rodberg
- Department of Biology, University of Massachusetts Amherst, Amherst, MA, USA
- Neuroscience and Behavior Program, University of Massachusetts Amherst, Amherst, MA, USA
| | - Ehsan Arabzadeh
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, The Australian National University, Canberra, AUS
| | - Chloe J. Bair-Marshall
- Neuroscience Institute, NYU Langone Medical Center, New York University, New York, New York, USA
| | - Craig W. Berridge
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, USA
| | - Esther Berrocoso
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, School of Medicine, Biomedical Research and Innovation Institute of Cádiz (INiBICA), University of Cadiz, Cadiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | | | | | - Alicia Che
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
| | - Oscar Davy
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | | | - Anthony M. Downs
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gabrielle Drummond
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Roman Dvorkin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Zeinab Fazlali
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
- Department of Psychiatry, Columbia University, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Robert C. Froemke
- Neuroscience Institute, NYU Langone Medical Center, New York University, New York, New York, USA
- Department of Otolaryngology, NYU Grossman School of Medicine, New York, NY, USA
| | - Erin Glennon
- Neuroscience Institute, NYU Langone Medical Center, New York University, New York, New York, USA
- Department of Neurology, Weill Cornell Medicine, New York
| | - Joshua I. Gold
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Hiroki Ito
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
- Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Xiaolong Jiang
- Department of Neuroscience, Baylor College of Medicine Neurological Research Institute at Texas Children’s Hospital, 1250, Houston, TX, USA
- Department of Ophthalmology, Baylor College of Medicine Neurological Research Institute at Texas Children’s Hospital, 1250, Houston, TX, USA
| | | | - Alfred P. Kaye
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
- Clinical Neurosciences Division, VA National Center for PTSD, West Haven, CT, USA
| | - Jenny R. Kim
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Chao-Cheng Kuo
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Rong-Jian Liu
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Yang Liu
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Meritxell Llorca-Torralba
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, School of Medicine, Biomedical Research and Innovation Institute of Cádiz (INiBICA), University of Cadiz, Cadiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Jordan G. McCall
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Zoe A. McElligott
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew M. McKinney
- Department of Neuroscience, Baylor College of Medicine Neurological Research Institute at Texas Children’s Hospital, 1250, Houston, TX, USA
| | - Cristina Miguelez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Ming-Yuan Min
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Alexandra C. Nowlan
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mohsen Omrani
- Department of Psychiatry, Queen’s University, Kingston, ON, Canada
| | - Gina R. Poe
- Integrative Biology and Physiology, UCLA, Los Angeles, CA, USA
| | - Anthony Edward Pickering
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Yadollah Ranjbar-Slamloo
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Jone Razquin
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Charles Rodenkirch
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Anna C. Sales
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Rath Satyasambit
- RIKEN Center for Brain Science, Wako-shi Saitama, Japan
- Department of Computer Science, Tokyo Institute of Technology, Midori, Yokohama, Japan
| | | | - Mriganka Sur
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Sonia Torres-Sanchez
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, School of Medicine, Biomedical Research and Innovation Institute of Cádiz (INiBICA), University of Cadiz, Cadiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Akira Uematsu
- Human Informatics and Information Research Institute, National Institute of Advanced Industrial Science and Technology, Japan
| | - Chayla R. Vazquez
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Amelien Vreven
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Qi Wang
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | | | - Hsiu-Wen Yang
- Department of Biomedical Sciences, Chung-Shan Medical University, Taichung, Taiwan
| | - Jen-Hau Yang
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Doctoral Program of Clinical and Experimental Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Liping Zhao
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA USA
| | - Ioannis S. Zouridis
- Graduate Training Centre of Neuroscience, International Max Planck Research School (IMPRS), University of Tübingen, Tübingen, Germany
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | | | - Elena Vazey
- Department of Biology, University of Massachusetts Amherst, Amherst, MA, USA
- Neuroscience and Behavior Program, University of Massachusetts Amherst, Amherst, MA, USA
| | - Nelson K. Totah
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| |
Collapse
|
8
|
Zhang S, Lu J, Jin Z, Xu H, Zhang D, Chen J, Wang J. Gut microbiota metabolites: potential therapeutic targets for Alzheimer's disease? Front Pharmacol 2024; 15:1459655. [PMID: 39355779 PMCID: PMC11442227 DOI: 10.3389/fphar.2024.1459655] [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: 07/04/2024] [Accepted: 09/05/2024] [Indexed: 10/03/2024] Open
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive decline in cognitive function, which significantly increases pain and social burden. However, few therapeutic interventions are effective in preventing or mitigating the progression of AD. An increasing number of recent studies support the hypothesis that the gut microbiome and its metabolites may be associated with upstream regulators of AD pathology. Methods In this review, we comprehensively explore the potential mechanisms and currently available interventions targeting the microbiome for the improvement of AD. Our discussion is structured around modern research advancements in AD, the bidirectional communication between the gut and brain, the multi-target regulatory effects of microbial metabolites on AD, and therapeutic strategies aimed at modulating gut microbiota to manage AD. Results The gut microbiota plays a crucial role in the pathogenesis of AD through continuous bidirectional communication via the microbiota-gut-brain axis. Among these, microbial metabolites such as lipids, amino acids, bile acids and neurotransmitters, especially sphingolipids and phospholipids, may serve as central components of the gut-brain axis, regulating AD-related pathogenic mechanisms including β-amyloid metabolism, Tau protein phosphorylation, and neuroinflammation. Additionally, interventions such as probiotic administration, fecal microbiota transplantation, and antibiotic use have also provided evidence supporting the association between gut microbiota and AD. At the same time, we propose an innovative strategy for treating AD: a healthy lifestyle combined with targeted probiotics and other potential therapeutic interventions, aiming to restore intestinal ecology and microbiota balance. Conclusion Despite previous efforts, the molecular mechanisms by which gut microbes act on AD have yet to be fully described. However, intestinal microorganisms may become an essential target for connecting the gut-brain axis and improving the symptoms of AD. At the same time, it requires joint exploration by multiple centers and multiple disciplines.
Collapse
Affiliation(s)
- Shanshan Zhang
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Jing Lu
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Ziqi Jin
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Hanying Xu
- Department of Encephalopathy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Dongmei Zhang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Jianan Chen
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Jian Wang
- Department of Encephalopathy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
9
|
Iannitelli AF, Hassenein L, Mulvey B, Blankenship HE, Liles LC, Sharpe AL, Pare JF, Segal A, Sloan SA, Martinowich K, McCann KE, Dougherty JD, Smith Y, Beckstead MJ, Weinshenker D. Tyrosinase-induced neuromelanin accumulation triggers rapid dysregulation and degeneration of the mouse locus coeruleus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.07.530845. [PMID: 36945637 PMCID: PMC10028911 DOI: 10.1101/2023.03.07.530845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
The locus coeruleus (LC), the major source of norepinephrine (NE) in the brain, is an early site of pathology in both Alzheimer's disease (AD) and Parkinson's disease (PD), and it undergoes catastrophic degeneration later in both disorders. Dysregulation of the LC is thought to contribute to prodromal symptoms of AD and PD such as anxiety and sleep disturbances, while frank LC-NE loss promotes cognitive decline. However, the mechanisms responsible for its selective vulnerability are unknown. The LC is among the only structures in the brain that produces appreciable amounts of neuromelanin (NM), a dark cytoplasmic pigment. It has been proposed that NM initially plays a protective role by sequestering toxic catecholamine metabolites and heavy metals, but may become harmful during aging as it overwhelms cellular machinery and is released during neurodegeneration. Rodents do not naturally produce NM, limiting the study of causal relationships between NM and LC pathology. Adapting a viral-mediated approach for expression of human tyrosinase, the enzyme responsible for peripheral melanin production, we successfully promoted pigmentation in mouse LC neurons that recapitulates key ultrastructural features of endogenous NM found in primates. Pigment expression results in LC neuron hyperactivity, reduced tissue NE levels, transcriptional changes, and novelty-induced anxiety phenotypes as early as 1-week post-injection. By 6-10 weeks, NM accumulation is associated with severe LC neuron neurodegeneration and microglial engulfment of the pigment granules, while the anxiety-like behavior is abated. These phenotypes are reminiscent of LC dysfunction and cell death in AD and PD, validating this model for studying the consequences of pigment accumulation in the LC as it relates to neurodegenerative disease.
Collapse
Affiliation(s)
- Alexa F. Iannitelli
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Leslie Hassenein
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Bernard Mulvey
- Department of Psychiatry, Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Harris E. Blankenship
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - L. Cameron Liles
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Amanda L. Sharpe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, 73117
| | - Jean-Francoise Pare
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Arielle Segal
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Steven A. Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Katharine E. McCann
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Joseph D. Dougherty
- Department of Psychiatry, Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yoland Smith
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Michael J. Beckstead
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
10
|
Zhang K, Zhu Y, Fenik P, Fleysh D, Ly C, Thomas SA, Veasey S. Norepinephrine Drives Sleep Fragmentation Activation of Asparagine Endopeptidase, Locus Ceruleus Degeneration, and Hippocampal Amyloid-β 42 Accumulation. J Neurosci 2024; 44:e1929232024. [PMID: 38830763 PMCID: PMC11236578 DOI: 10.1523/jneurosci.1929-23.2024] [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: 10/11/2023] [Revised: 03/20/2024] [Accepted: 04/11/2024] [Indexed: 06/05/2024] Open
Abstract
Chronic sleep disruption (CSD), from insufficient or fragmented sleep and is an important risk factor for Alzheimer's disease (AD). Underlying mechanisms are not understood. CSD in mice results in degeneration of locus ceruleus neurons (LCn) and CA1 hippocampal neurons and increases hippocampal amyloid-β42 (Aβ42), entorhinal cortex (EC) tau phosphorylation (p-tau), and glial reactivity. LCn injury is increasingly implicated in AD pathogenesis. CSD increases NE turnover in LCn, and LCn norepinephrine (NE) metabolism activates asparagine endopeptidase (AEP), an enzyme known to cleave amyloid precursor protein (APP) and tau into neurotoxic fragments. We hypothesized that CSD would activate LCn AEP in an NE-dependent manner to induce LCn and hippocampal injury. Here, we studied LCn, hippocampal, and EC responses to CSD in mice deficient in NE [dopamine β-hydroxylase (Dbh)-/-] and control male and female mice, using a model of chronic fragmentation of sleep (CFS). Sleep was equally fragmented in Dbh -/- and control male and female mice, yet only Dbh -/- mice conferred resistance to CFS loss of LCn, LCn p-tau, and LCn AEP upregulation and activation as evidenced by an increase in AEP-cleaved APP and tau fragments. Absence of NE also prevented a CFS increase in hippocampal AEP-APP and Aβ42 but did not prevent CFS-increased AEP-tau and p-tau in the EC. Collectively, this work demonstrates AEP activation by CFS, establishes key roles for NE in both CFS degeneration of LCn neurons and CFS promotion of forebrain Aβ accumulation, and, thereby, identifies a key molecular link between CSD and specific AD neural injuries.
Collapse
Affiliation(s)
- Kathy Zhang
- Departments of Medicine and Pharmacology, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Yan Zhu
- Departments of Medicine and Pharmacology, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Polina Fenik
- Departments of Medicine and Pharmacology, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Dennis Fleysh
- Departments of Medicine and Pharmacology, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Colin Ly
- Departments of Medicine and Pharmacology, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Steven A Thomas
- Departments of Medicine and Pharmacology, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Sigrid Veasey
- Departments of Medicine and Pharmacology, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
11
|
Engels-Domínguez N, Koops EA, Hsieh S, Wiklund EE, Schultz AP, Riphagen JM, Prokopiou PC, Hanseeuw BJ, Rentz DM, Sperling RA, Johnson KA, Jacobs HIL. Lower in vivo locus coeruleus integrity is associated with lower cortical thickness in older individuals with elevated Alzheimer's pathology: a cohort study. Alzheimers Res Ther 2024; 16:129. [PMID: 38886798 PMCID: PMC11181564 DOI: 10.1186/s13195-024-01500-0] [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: 11/30/2023] [Accepted: 06/12/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Autopsy work indicates that the widely-projecting noradrenergic pontine locus coeruleus (LC) is among the earliest regions to accumulate hyperphosphorylated tau, a neuropathological Alzheimer's disease (AD) hallmark. This early tau deposition is accompanied by a reduced density of LC projections and a reduction of norepinephrine's neuroprotective effects, potentially compromising the neuronal integrity of LC's cortical targets. Previous studies suggest that lower magnetic resonance imaging (MRI)-derived LC integrity may signal cortical tissue degeneration in cognitively healthy, older individuals. However, whether these observations are driven by underlying AD pathology remains unknown. To that end, we examined potential effect modifications by cortical beta-amyloid and tau pathology on the association between in vivo LC integrity, as quantified by LC MRI signal intensity, and cortical neurodegeneration, as indexed by cortical thickness. METHODS A total of 165 older individuals (74.24 ± 9.72 years, ~ 60% female, 10% cognitively impaired) underwent whole-brain and dedicated LC 3T-MRI, Pittsburgh Compound-B (PiB, beta-amyloid) and Flortaucipir (FTP, tau) positron emission tomography. Linear regression analyses with bootstrapped standard errors (n = 2000) assessed associations between bilateral cortical thickness and i) LC MRI signal intensity and, ii) LC MRI signal intensity interacted with cortical FTP or PiB (i.e., EC FTP, IT FTP, neocortical PiB) in the entire sample and a low beta-amyloid subsample. RESULTS Across the entire sample, we found a direct effect, where lower LC MRI signal intensity was associated with lower mediolateral temporal cortical thickness. Evaluation of potential effect modifications by FTP or PiB revealed that lower LC MRI signal intensity was related to lower cortical thickness, particularly in individuals with elevated (EC, IT) FTP or (neocortical) PiB. The latter result was present starting from subthreshold PiB values. In low PiB individuals, lower LC MRI signal intensity was related to lower EC cortical thickness in the context of elevated EC FTP. CONCLUSIONS Our findings suggest that LC-related cortical neurodegeneration patterns in older individuals correspond to regions representing early Braak stages and may reflect a combination of LC projection density loss and emergence of cortical AD pathology. This provides a novel understanding that LC-related cortical neurodegeneration may signal downstream consequences of AD-related pathology, rather than being exclusively a result of aging.
Collapse
Affiliation(s)
- Nina Engels-Domínguez
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
| | - Elouise A Koops
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Stephanie Hsieh
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Emma E Wiklund
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Aaron P Schultz
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joost M Riphagen
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Prokopis C Prokopiou
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Bernard J Hanseeuw
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Dorene M Rentz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Keith A Johnson
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Heidi I L Jacobs
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA.
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
12
|
Martin SC, Joyce KK, Lord JS, Harper KM, Nikolova VD, Cohen TJ, Moy SS, Diering GH. Sleep Disruption Precedes Forebrain Synaptic Tau Burden and Contributes to Cognitive Decline in a Sex-Dependent Manner in the P301S Tau Transgenic Mouse Model. eNeuro 2024; 11:ENEURO.0004-24.2024. [PMID: 38858068 PMCID: PMC11209651 DOI: 10.1523/eneuro.0004-24.2024] [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: 01/02/2024] [Revised: 05/02/2024] [Accepted: 05/14/2024] [Indexed: 06/12/2024] Open
Abstract
Sleep disruption and impaired synaptic processes are common features in neurodegenerative diseases, including Alzheimer's disease (AD). Hyperphosphorylated Tau is known to accumulate at neuronal synapses in AD, contributing to synapse dysfunction. However, it remains unclear how sleep disruption and synapse pathology interact to contribute to cognitive decline. Here, we examined sex-specific onset and consequences of sleep loss in AD/tauopathy model PS19 mice. Using a piezoelectric home-cage monitoring system, we showed PS19 mice exhibited early-onset and progressive hyperarousal, a selective dark-phase sleep disruption, apparent at 3 months in females and 6 months in males. Using the Morris water maze test, we report that chronic sleep disruption (CSD) accelerated the onset of decline of hippocampal spatial memory in PS19 males only. Hyperarousal occurs well in advance of robust forebrain synaptic Tau burden that becomes apparent at 6-9 months. To determine whether a causal link exists between sleep disruption and synaptic Tau hyperphosphorylation, we examined the correlation between sleep behavior and synaptic Tau, or exposed mice to acute or chronic sleep disruption at 6 months. While we confirm that sleep disruption is a driver of Tau hyperphosphorylation in neurons of the locus ceruleus, we were unable to show any causal link between sleep loss and Tau burden in forebrain synapses. Despite the finding that hyperarousal appears earlier in females, female cognition was resilient to the effects of sleep disruption. We conclude sleep disruption interacts with the synaptic Tau burden to accelerate the onset of cognitive decline with greater vulnerability in males.
Collapse
Affiliation(s)
- Shenée C Martin
- Departments of Cell Biology and Physiology, Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Kathryn K Joyce
- Departments of Cell Biology and Physiology, Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Julia S Lord
- Departments of Cell Biology and Physiology, Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Kathryn M Harper
- Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Viktoriya D Nikolova
- Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Todd J Cohen
- Neurology, Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Sheryl S Moy
- Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
- Carolina Institute for Developmental Disabilities, Carrboro, North Carolina 27510
| | - Graham H Diering
- Departments of Cell Biology and Physiology, Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
- Carolina Institute for Developmental Disabilities, Carrboro, North Carolina 27510
| |
Collapse
|
13
|
Pahl J, Prokopiou PC, Bueichekú E, Schultz AP, Papp KV, Farrell ME, Rentz DM, Sperling RA, Johnson KA, Jacobs HIL. Locus coeruleus integrity and left frontoparietal connectivity provide resilience against attentional decline in preclinical alzheimer's disease. Alzheimers Res Ther 2024; 16:119. [PMID: 38822365 PMCID: PMC11140954 DOI: 10.1186/s13195-024-01485-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/22/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND Autopsy work reported that neuronal density in the locus coeruleus (LC) provides neural reserve against cognitive decline in dementia. Recent neuroimaging and pharmacological studies reported that left frontoparietal network functional connectivity (LFPN-FC) confers resilience against beta-amyloid (Aβ)-related cognitive decline in preclinical sporadic and autosomal dominant Alzheimer's disease (AD), as well as against LC-related cognitive changes. Given that the LFPN and the LC play important roles in attention, and attention deficits have been observed early in the disease process, we examined whether LFPN-FC and LC structural health attenuate attentional decline in the context of AD pathology. METHODS 142 participants from the Harvard Aging Brain Study who underwent resting-state functional MRI, LC structural imaging, PiB(Aβ)-PET, and up to 5 years of cognitive follow-ups were included (mean age = 74.5 ± 9.9 years, 89 women). Cross-sectional robust linear regression associated LC integrity (measured as the average of five continuous voxels with the highest intensities in the structural LC images) or LFPN-FC with Digit Symbol Substitution Test (DSST) performance at baseline. Longitudinal robust mixed effect analyses examined associations between DSST decline and (i) two-way interactions of baseline LC integrity (or LFPN-FC) and PiB or (ii) the three-way interaction of baseline LC integrity, LFPN-FC, and PiB. Baseline age, sex, and years of education were included as covariates. RESULTS At baseline, lower LFPN-FC, but not LC integrity, was related to worse DSST performance. Longitudinally, lower baseline LC integrity was associated with a faster DSST decline, especially at PiB > 10.38 CL. Lower baseline LFPN-FC was associated with a steeper decline on the DSST but independent of PiB. At elevated PiB levels (> 46 CL), higher baseline LFPN-FC was associated with an attenuated decline on the DSST, despite the presence of lower LC integrity. CONCLUSIONS Our findings demonstrate that the LC can provide resilience against Aβ-related attention decline. However, when Aβ accumulates and the LC's resources may be depleted, the functioning of cortical target regions of the LC, such as the LFPN-FC, can provide additional resilience to sustain attentional performance in preclinical AD. These results provide critical insights into the neural correlates contributing to individual variability at risk versus resilience against Aβ-related cognitive decline.
Collapse
Affiliation(s)
- Jennifer Pahl
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Prokopis C Prokopiou
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Elisenda Bueichekú
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathryn V Papp
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michelle E Farrell
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dorene M Rentz
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Reisa A Sperling
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Keith A Johnson
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Heidi I L Jacobs
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Bueichekú E, Diez I, Kim CM, Becker JA, Koops EA, Kwong K, Papp KV, Salat DH, Bennett DA, Rentz DM, Sperling RA, Johnson KA, Sepulcre J, Jacobs HIL. Spatiotemporal patterns of locus coeruleus integrity predict cortical tau and cognition. NATURE AGING 2024; 4:625-637. [PMID: 38664576 PMCID: PMC11108787 DOI: 10.1038/s43587-024-00626-y] [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: 04/11/2023] [Accepted: 04/04/2024] [Indexed: 04/28/2024]
Abstract
Autopsy studies indicated that the locus coeruleus (LC) accumulates hyperphosphorylated tau before allocortical regions in Alzheimer's disease. By combining in vivo longitudinal magnetic resonance imaging measures of LC integrity, tau positron emission tomography imaging and cognition with autopsy data and transcriptomic information, we examined whether LC changes precede allocortical tau deposition and whether specific genetic features underlie LC's selective vulnerability to tau. We found that LC integrity changes preceded medial temporal lobe tau accumulation, and together these processes were associated with lower cognitive performance. Common gene expression profiles between LC-medial temporal lobe-limbic regions map to biological functions in protein transport regulation. These findings advance our understanding of the spatiotemporal patterns of initial tau spreading from the LC and LC's selective vulnerability to Alzheimer's disease pathology. LC integrity measures can be a promising indicator for identifying the time window when individuals are at risk of disease progression and underscore the importance of interventions mitigating initial tau spread.
Collapse
Affiliation(s)
- Elisenda Bueichekú
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Ibai Diez
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Chan-Mi Kim
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - John Alex Becker
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Kenneth Kwong
- Harvard Medical School, Boston, MA, USA
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Kathryn V Papp
- Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - David H Salat
- Harvard Medical School, Boston, MA, USA
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Neuroimaging Research for Veterans Center, VA Boston Healthcare System, Boston, MA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Dorene M Rentz
- Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Reisa A Sperling
- Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Keith A Johnson
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Jorge Sepulcre
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA.
- Department of Radiology, Yale PET Center, Yale Medical School, Yale University, New Haven, CT, USA.
| | - Heidi I L Jacobs
- Harvard Medical School, Boston, MA, USA.
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA.
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, Netherlands.
| |
Collapse
|
15
|
Evans AK, Saw NL, Woods CE, Vidano LM, Blumenfeld SE, Lam RK, Chu EK, Reading C, Shamloo M. Impact of high-fat diet on cognitive behavior and central and systemic inflammation with aging and sex differences in mice. Brain Behav Immun 2024; 118:334-354. [PMID: 38408498 PMCID: PMC11019935 DOI: 10.1016/j.bbi.2024.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 02/28/2024] Open
Abstract
Aging and age-related diseases are associated with cellular stress, metabolic imbalance, oxidative stress, and neuroinflammation, accompanied by cognitive impairment. Lifestyle factors such as diet, sleep fragmentation, and stress can potentiate damaging cellular cascades and lead to an acceleration of brain aging and cognitive impairment. High-fat diet (HFD) has been associated with obesity, metabolic disorders like diabetes, and cardiovascular disease. HFD also induces neuroinflammation, impairs learning and memory, and may increase anxiety-like behavior. Effects of a HFD may also vary between sexes. The interaction between Age- and Sex- and Diet-related changes in neuroinflammation and cognitive function is an important and poorly understood area of research. This study was designed to examine the effects of HFD on neuroinflammation, behavior, and neurodegeneration in mice in the context of aging or sex differences. In a series of studies, young (2-3 months) or old (12-13 months) C57BL/6J male mice or young male and female C57Bl/6J mice were fed either a standard diet (SD) or a HFD for 5-6 months. Behavior was assessed in Activity Chamber, Y-maze, Novel Place Recognition, Novel Object Recognition, Elevated Plus Maze, Open Field, Morris Water Maze, and Fear Conditioning. Post-mortem analyses assessed a panel of inflammatory markers in the plasma and hippocampus. Additionally, proteomic analysis of the hypothalamus, neurodegeneration, neuroinflammation in the locus coeruleus, and neuroinflammation in the hippocampus were assessed in a subset of young and aged male mice. We show that HFD increased body weight and decreased locomotor activity across groups compared to control mice fed a SD. HFD altered anxiety-related exploratory behavior. HFD impaired spatial learning and recall in young male mice and impaired recall in cued fear conditioning in young and aged male mice, with no effects on spatial learning or fear conditioning in young female mice. Effects of Age and Sex were observed on neuroinflammatory cytokines, with only limited effects of HFD. HFD had a more significant impact on systemic inflammation in plasma across age and sex. Aged male mice had induction of microglial immunoreactivity in both the locus coeruleus (LC) and hippocampus an effect that HFD exacerbated in the hippocampal CA1 region. Proteomic analysis of the hypothalamus revealed changes in pathways related to metabolism and neurodegeneration with both aging and HFD in male mice. Our findings suggest that HFD induces widespread systemic inflammation and limited neuroinflammation. In addition, HFD alters exploratory behavior in male and female mice, and impairs learning and memory in male mice. These results provide valuable insight into the impact of diet on cognition and aging pathophysiology.
Collapse
Affiliation(s)
- Andrew K Evans
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | - Nay L Saw
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | - Claire E Woods
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | - Laura M Vidano
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | - Sarah E Blumenfeld
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | - Rachel K Lam
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | - Emily K Chu
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | | | - Mehrdad Shamloo
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304.
| |
Collapse
|
16
|
Tansey MG, Boles J, Holt J, Cole C, Neighbarger N, Urs N, Uriarte-Huarte O. Locus coeruleus injury modulates ventral midbrain neuroinflammation during DSS-induced colitis. RESEARCH SQUARE 2024:rs.3.rs-3952442. [PMID: 38559083 PMCID: PMC10980147 DOI: 10.21203/rs.3.rs-3952442/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Parkinson's disease (PD) is characterized by a decades-long prodrome, consisting of a collection of non-motor symptoms that emerges prior to the motor manifestation of the disease. Of these non-motor symptoms, gastrointestinal dysfunction and deficits attributed to central norepinephrine (NE) loss, including mood changes and sleep disturbances, are frequent in the PD population and emerge early in the disease. Evidence is mounting that injury and inflammation in the gut and locus coeruleus (LC), respectively, underlie these symptoms, and the injury of these systems is central to the progression of PD. In this study, we generate a novel two-hit mouse model that captures both features, using dextran sulfate sodium (DSS) to induce gut inflammation and N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4) to lesion the LC. We first confirmed the specificity of DSP-4 for central NE using neurochemical methods and fluorescence light-sheet microscopy of cleared tissue, and established that DSS-induced outcomes in the periphery, including weight loss, gross indices of gut injury and systemic inflammation, the loss of tight junction proteins in the colonic epithelium, and markers of colonic inflammation, were unaffected with DSP-4 pre-administration. We then measured alterations in neuroimmune gene expression in the ventral midbrain in response to DSS treatment alone as well as the extent to which prior LC injury modified this response. In this two-hit model we observed that DSS-induced colitis activates the expression of key cytokines and chemokines in the ventral midbrain only in the presence of LC injury and the typical DSS-associated neuroimmune is blunted by pre-LC lesioning with DSP-4. In all, this study supports the growing appreciation for the LC as neuroprotective against inflammation-induced brain injury and draws attention to the potential for NEergic interventions to exert disease-modifying effects under conditions where peripheral inflammation may compromise ventral midbrain dopaminergic neurons and increase the risk for development of PD.
Collapse
|
17
|
Martin SC, Joyce KK, Harper KM, Harp SJ, Cohen TJ, Moy SS, Diering GH. Evaluating Fatty Acid Amide Hydrolase as a Suitable Target for Sleep Promotion in a Transgenic TauP301S Mouse Model of Neurodegeneration. Pharmaceuticals (Basel) 2024; 17:319. [PMID: 38543105 PMCID: PMC10975243 DOI: 10.3390/ph17030319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 04/01/2024] Open
Abstract
Sleep disruption is an expected component of aging and neurodegenerative conditions, including Alzheimer's disease (AD). Sleep disruption has been demonstrated as a driver of AD pathology and cognitive decline. Therefore, treatments designed to maintain sleep may be effective in slowing or halting AD progression. However, commonly used sleep aid medications are associated with an increased risk of AD, highlighting the need for sleep aids with novel mechanisms of action. The endocannabinoid system holds promise as a potentially effective and novel sleep-enhancing target. By using pharmacology and genetic knockout strategies, we evaluated fatty acid amide hydrolase (FAAH) as a therapeutic target to improve sleep and halt disease progression in a transgenic Tau P301S (PS19) model of Tauopathy and AD. We have recently shown that PS19 mice exhibit sleep disruption in the form of dark phase hyperarousal as an early symptom that precedes robust Tau pathology and cognitive decline. Acute FAAH inhibition with PF3845 resulted in immediate improvements in sleep behaviors in male and female PS19 mice, supporting FAAH as a potentially suitable sleep-promoting target. Moreover, sustained drug dosing for 5-10 days resulted in maintained improvements in sleep. To evaluate the effect of chronic FAAH inhibition as a possible therapeutic strategy, we generated FAAH-/- PS19 mice models. Counter to our expectations, FAAH knockout did not protect PS19 mice from progressive sleep loss, neuroinflammation, or cognitive decline. Our results provide support for FAAH as a novel target for sleep-promoting therapies but further indicate that the complete loss of FAAH activity may be detrimental.
Collapse
Affiliation(s)
- Shenée C. Martin
- Department of Cell Biology and Physiology and the Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kathryn K. Joyce
- Department of Cell Biology and Physiology and the Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kathryn M. Harper
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Samuel J. Harp
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Todd J. Cohen
- Department of Neurology and the Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sheryl S. Moy
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Carolina Institute for Developmental Disabilities, Carrboro, NC 27510, USA
| | - Graham H. Diering
- Department of Cell Biology and Physiology and the Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Carolina Institute for Developmental Disabilities, Carrboro, NC 27510, USA
| |
Collapse
|
18
|
Granholm AC, Hamlett ED. The Role of Tau Pathology in Alzheimer's Disease and Down Syndrome. J Clin Med 2024; 13:1338. [PMID: 38592182 PMCID: PMC10932364 DOI: 10.3390/jcm13051338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/10/2024] [Accepted: 02/20/2024] [Indexed: 04/10/2024] Open
Abstract
Background: Individuals with Down syndrome (DS) exhibit an almost complete penetrance of Alzheimer's disease (AD) pathology but are underrepresented in clinical trials for AD. The Tau protein is associated with microtubule function in the neuron and is crucial for normal axonal transport. In several different neurodegenerative disorders, Tau misfolding leads to hyper-phosphorylation of Tau (p-Tau), which may seed pathology to bystander cells and spread. This review is focused on current findings regarding p-Tau and its potential to seed pathology as a "prion-like" spreader. It also considers the consequences of p-Tau pathology leading to AD, particularly in individuals with Down syndrome. Methods: Scopus (SC) and PubMed (PM) were searched in English using keywords "tau AND seeding AND brain AND down syndrome". A total of 558 SC or 529 PM potentially relevant articles were identified, of which only six SC or three PM articles mentioned Down syndrome. This review was built upon the literature and the recent findings of our group and others. Results: Misfolded p-Tau isoforms are seeding competent and may be responsible for spreading AD pathology. Conclusions: This review demonstrates recent work focused on understanding the role of neurofibrillary tangles and monomeric/oligomeric Tau in the prion-like spreading of Tau pathology in the human brain.
Collapse
Affiliation(s)
- Ann-Charlotte Granholm
- Department of Neurosurgery, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA
| | - Eric D. Hamlett
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
| |
Collapse
|
19
|
Boles JS, Holt J, Cole CL, Neighbarger NK, Urs NM, Huarte OU, Tansey MG. Locus coeruleus injury modulates ventral midbrain neuroinflammation during DSS-induced colitis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.12.580010. [PMID: 38405709 PMCID: PMC10888767 DOI: 10.1101/2024.02.12.580010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Parkinson's disease (PD) is characterized by a decades-long prodrome, consisting of a collection of non-motor symptoms that emerges prior to the motor manifestation of the disease. Of these non-motor symptoms, gastrointestinal dysfunction and deficits attributed to central norepinephrine (NE) loss, including mood changes and sleep disturbances, are frequent in the PD population and emerge early in the disease. Evidence is mounting that injury and inflammation in the gut and locus coeruleus (LC), respectively, underlie these symptoms, and the injury of these systems is central to the progression of PD. In this study, we generate a novel two-hit mouse model that captures both features, using dextran sulfate sodium (DSS) to induce gut inflammation and N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4) to lesion the LC. We first confirmed the specificity of DSP-4 for central NE using neurochemical methods and fluorescence light-sheet microscopy of cleared tissue, and established that DSS-induced outcomes in the periphery, including weight loss, gross indices of gut injury and systemic inflammation, the loss of tight junction proteins in the colonic epithelium, and markers of colonic inflammation, were unaffected with DSP-4 pre-administration. We then measured alterations in neuroimmune gene expression in the ventral midbrain in response to DSS treatment alone as well as the extent to which prior LC injury modified this response. In this two-hit model we observed that DSS-induced colitis activates the expression of key cytokines and chemokines in the ventral midbrain only in the presence of LC injury and the typical DSS-associated neuroimmune is blunted by pre-LC lesioning with DSP-4. In all, this study supports the growing appreciation for the LC as neuroprotective against inflammation-induced brain injury and draws attention to the potential for NEergic interventions to exert disease-modifying effects under conditions where peripheral inflammation may compromise ventral midbrain dopaminergic neurons and increase the risk for development of PD.
Collapse
Affiliation(s)
- Jake Sondag Boles
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Jenny Holt
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Cassandra L. Cole
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Noelle K. Neighbarger
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Nikhil M. Urs
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Department of Pharmacology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Oihane Uriarte Huarte
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Malú Gámez Tansey
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
20
|
Liu R, Guo Z, Li M, Liu S, Zhi Y, Jiang Z, Liang X, Hu H, Zhu J. Lower fractional dimension in Alzheimer's disease correlates with reduced locus coeruleus signal intensity. Magn Reson Imaging 2024; 106:24-30. [PMID: 37541457 DOI: 10.1016/j.mri.2023.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/19/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023]
Abstract
This study aimed to determine the pattern of fractional dimension (FD) in Alzheimer's disease (AD) patients, and investigate the relationship between FD and the locus coeruleus (LC) signal intensity.A total of 27 patients with AD and 25 healthy controls (HC) were collected to estimate the pattern of fractional dimension (FD) and cortical thickness (CT) using the Computational Anatomy Toolbox (CAT12), and statistically analyze between groups on a vertex level using statistical parametric mapping 12. In addition, they were examined by neuromelanin sensitive MRI(NM-MRI) technique to calculate the locus coeruleus signal contrast ratios (LC-CRs). Additionally, correlations between the pattern of FD and LC-CRs were further examined.Compared to HC, AD patients showed widespread lower CT and FD Furthermore, significant positive correlation was found between local fractional dimension (LFD) of the left rostral middle frontal cortex and LC-CRs. Results suggest lower cortical LFD is associated with LCCRs that may reflect a reduction due to broader neurodegenerative processes. This finding may highlight the potential utility for advanced measures of cortical complexity in assessing brain health and early identification of neurodegenerative processes.
Collapse
Affiliation(s)
- Rong Liu
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215004, China
| | - Zhiwen Guo
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215004, China
| | - Meng Li
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215004, China
| | - Shanwen Liu
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215004, China
| | - Yuqi Zhi
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215004, China
| | - Zhen Jiang
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215004, China
| | - Xiaoyun Liang
- Institute of Artificial Intelligence and Clinical Innovation, Neusoft Medical Systems Co., Ltd., Shanghai 200241, China; Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC 3084, Australia
| | - Hua Hu
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215004, China.
| | - Jiangtao Zhu
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215004, China.
| |
Collapse
|
21
|
Matt RA, Martin RS, Evans AK, Gever JR, Vargas GA, Shamloo M, Ford AP. Locus Coeruleus and Noradrenergic Pharmacology in Neurodegenerative Disease. Handb Exp Pharmacol 2024; 285:555-616. [PMID: 37495851 DOI: 10.1007/164_2023_677] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Adrenoceptors (ARs) throughout the brain are stimulated by noradrenaline originating mostly from neurons of the locus coeruleus, a brainstem nucleus that is ostensibly the earliest to show detectable pathology in neurodegenerative diseases such as Alzheimer's and Parkinson's diseases. The α1-AR, α2-AR, and β-AR subtypes expressed in target brain regions and on a range of cell populations define the physiological responses to noradrenaline, which includes activation of cognitive function in addition to modulation of neurometabolism, cerebral blood flow, and neuroinflammation. As these heterocellular functions are critical for maintaining brain homeostasis and neuronal health, combating the loss of noradrenergic tone from locus coeruleus degeneration may therefore be an effective treatment for both cognitive symptoms and disease modification in neurodegenerative indications. Two pharmacologic approaches are receiving attention in recent clinical studies: preserving noradrenaline levels (e.g., via reuptake inhibition) and direct activation of target adrenoceptors. Here, we review the expression and role of adrenoceptors in the brain, the preclinical studies which demonstrate that adrenergic stimulation can support cognitive function and cerebral health by reversing the effects of noradrenaline depletion, and the human data provided by pharmacoepidemiologic analyses and clinical trials which together identify adrenoceptors as promising targets for the treatment of neurodegenerative disease.
Collapse
Affiliation(s)
| | | | - Andrew K Evans
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | | | | | - Mehrdad Shamloo
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | | |
Collapse
|
22
|
Galgani A, Giorgi FS. Exploring the Role of Locus Coeruleus in Alzheimer's Disease: a Comprehensive Update on MRI Studies and Implications. Curr Neurol Neurosci Rep 2023; 23:925-936. [PMID: 38064152 PMCID: PMC10724305 DOI: 10.1007/s11910-023-01324-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2023] [Indexed: 12/18/2023]
Abstract
PURPOSE OF REVIEW Performing a thorough review of magnetic resonance imaging (MRI) studies assessing locus coeruleus (LC) integrity in ageing and Alzheimer's disease (AD), and contextualizing them with current preclinical and neuropathological literature. RECENT FINDINGS MRI successfully detected LC alterations in ageing and AD, identifying degenerative phenomena involving this nucleus even in the prodromal stages of the disorder. The degree of LC disruption was also associated with the severity of AD cortical pathology, cognitive and behavioral impairment, and the risk of clinical progression. Locus coeruleus-MRI has proved to be a useful tool to assess the integrity of the central noradrenergic system in vivo in humans. It allowed to test in patients preclinical and experimental hypothesis, thus confirming the specific and marked involvement of the LC in AD and its key pathogenetic role. Locus coeruleus-MRI-related data might represent the theoretical basis on which to start developing noradrenergic drugs to target AD.
Collapse
Affiliation(s)
- Alessandro Galgani
- Department of Translational Research and of New Surgical and Medical Technologies School of Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Filippo Sean Giorgi
- Department of Translational Research and of New Surgical and Medical Technologies School of Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy.
| |
Collapse
|
23
|
Gheidi A, Davidson CJ, Simpson SC, Yahya MA, Sadik N, Mascarin AT, Perrine SA. Norepinephrine depletion in the brain sex-dependently modulates aspects of spatial learning and memory in female and male rats. Psychopharmacology (Berl) 2023; 240:2585-2595. [PMID: 37658879 PMCID: PMC11069163 DOI: 10.1007/s00213-023-06453-0] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 08/22/2023] [Indexed: 09/05/2023]
Abstract
RATIONALE The contribution of norepinephrine on the different phases of spatial memory processing remains incompletely understood. To address this gap, this study depleted norepinephrine in the brain and then conducted a spatial learning task with multiple phases. METHODS Male and female Wistar rats were administered 50 mg/kg/i.p. of DSP-4 (N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine) to deplete norepinephrine. After 10 days, rats were trained on a 20-hole Barnes maze spatial navigation task for 5 days. On the fifth day, animals were euthanized and HPLC was used to confirm depletion of norepinephrine in select brain regions. In Experiment 2, rats underwent a similar Barnes maze procedure that continued beyond day 5 to investigate memory retrieval and updating via a single probe trial and two reversal learning periods. RESULTS Rats did not differ in Barnes maze acquisition between DSP-4 and saline-injected rats; however, initial acquisition differed between the sexes. HPLC analysis confirmed selective depletion of norepinephrine in dorsal hippocampus and cingulate cortex without impact to other monoamines. When retrieval was tested through a probe trial, DSP-4-improved memory retrieval in males but impaired it in females. Cognitive flexibility was transiently impacted by DSP-4 in males only. CONCLUSIONS Despite significantly reducing levels of norepinephrine, DSP-4 had only a modest impact on spatial learning and behavioral flexibility. Memory retrieval and early reversal learning were most affected and in a sex-specific manner. These data suggest that norepinephrine has sex-specific neuromodulatory effects on memory retrieval with a lesser effect on cognitive flexibility and no impact on acquisition of learned behavior.
Collapse
Affiliation(s)
- Ali Gheidi
- Department of Biomedical Sciences, Mercer University School of Medicine, 1550 College St., Macon, GA, 31207, USA.
| | - Cameron J Davidson
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Serena C Simpson
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Majd A Yahya
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nareen Sadik
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Alixandria T Mascarin
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Shane A Perrine
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
24
|
Krohn F, Lancini E, Ludwig M, Leiman M, Guruprasath G, Haag L, Panczyszyn J, Düzel E, Hämmerer D, Betts M. Noradrenergic neuromodulation in ageing and disease. Neurosci Biobehav Rev 2023; 152:105311. [PMID: 37437752 DOI: 10.1016/j.neubiorev.2023.105311] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/29/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023]
Abstract
The locus coeruleus (LC) is a small brainstem structure located in the lower pons and is the main source of noradrenaline (NA) in the brain. Via its phasic and tonic firing, it modulates cognition and autonomic functions and is involved in the brain's immune response. The extent of degeneration to the LC in healthy ageing remains unclear, however, noradrenergic dysfunction may contribute to the pathogenesis of Alzheimer's (AD) and Parkinson's disease (PD). Despite their differences in progression at later disease stages, the early involvement of the LC may lead to comparable behavioural symptoms such as preclinical sleep problems and neuropsychiatric symptoms as a result of AD and PD pathology. In this review, we draw attention to the mechanisms that underlie LC degeneration in ageing, AD and PD. We aim to motivate future research to investigate how early degeneration of the noradrenergic system may play a pivotal role in the pathogenesis of AD and PD which may also be relevant to other neurodegenerative diseases.
Collapse
Affiliation(s)
- F Krohn
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - E Lancini
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany.
| | - M Ludwig
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
| | - M Leiman
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - G Guruprasath
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - L Haag
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - J Panczyszyn
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - E Düzel
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neuroscience, University College London, London UK-WC1E 6BT, UK; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
| | - D Hämmerer
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neuroscience, University College London, London UK-WC1E 6BT, UK; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany; Department of Psychology, University of Innsbruck, A-6020 Innsbruck, Austria
| | - M Betts
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
| |
Collapse
|
25
|
Gargano A, Olabiyi BF, Palmisano M, Zimmer A, Bilkei-Gorzo A. Possible role of locus coeruleus neuronal loss in age-related memory and attention deficits. Front Neurosci 2023; 17:1264253. [PMID: 37694113 PMCID: PMC10492095 DOI: 10.3389/fnins.2023.1264253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Introduction Aging is associated with a decline in cognitive abilities, including memory and attention. It is generally accepted that age-related histological changes such as increased neuroinflammatory glial activity and a reduction in the number of specific neuronal populations contribute to cognitive aging. Noradrenergic neurons in the locus coeruleus (LC) undergo an approximately 20 % loss during ageing both in humans and mice, but whether this change contributes to cognitive deficits is not known. To address this issue, we asked whether a similar loss of LC neurons in young animals as observed in aged animals impairs memory and attention, cognitive domains that are both influenced by the noradrenergic system and impaired in aging. Methods For that, we treated young healthy mice with DSP-4, a toxin that specifically kills LC noradrenergic neurons. We compared the performance of DSP-4 treated young mice with the performance of aged mice in models of attention and memory. To do this, we first determined the dose of DSP-4, which causes a similar 20 % neuronal loss as is typical in aged animals. Results Young mice treated with DSP-4 showed impaired attention in the presence of distractor and memory deficits in the 5-choice serial reaction time test (5-CSRTT). Old, untreated mice showed severe deficits in both the 5-CSRTT and in fear extinction tests. Discussion Our data now suggest that a reduction in the number of LC neurons contributes to impaired working memory and greater distractibility in attentional tasks but not to deficits in fear extinction. We hypothesize that the moderate loss of LC noradrenergic neurons during aging contributes to attention deficits and working memory impairments.
Collapse
Affiliation(s)
| | | | | | | | - Andras Bilkei-Gorzo
- Medical Faculty, Institute of Molecular Psychiatry, University of Bonn, Venusberg-Campus, Bonn, Germany
| |
Collapse
|
26
|
Pilski A, Graves SM. Repeated Methamphetamine Administration Results in Axon Loss Prior to Somatic Loss of Substantia Nigra Pars Compacta and Locus Coeruleus Neurons in Male but Not Female Mice. Int J Mol Sci 2023; 24:13039. [PMID: 37685846 PMCID: PMC10487759 DOI: 10.3390/ijms241713039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/17/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Methamphetamine (meth) is a neurotoxic psychostimulant that increases monoamine oxidase (MAO)-dependent mitochondrial oxidant stress in axonal but not somatic compartments of substantia nigra pars compacta (SNc) and locus coeruleus (LC) neurons. Chronic meth administration results in the degeneration of SNc and LC neurons in male mice, and MAO inhibition is neuroprotective, suggesting that the deleterious effects of chronic meth begin in axons before advancing to the soma of SNc and LC neurons. To test this hypothesis, mice were administered meth (5 mg/kg) for 14, 21, or 28 days, and SNc and LC axonal lengths and numbers of neurons were quantified. In male mice, the SNc and LC axon lengths decreased with 14, 21, and 28 days of meth, whereas somatic loss was only observed after 28 days of meth; MAO inhibition (phenelzine; 20 mg/kg) prevented axonal and somatic loss of SNc and LC neurons. In contrast, chronic (28-day) meth had no effect on the axon length or numbers of SNc or LC neurons in female mice. The results demonstrate that repeated exposure to meth produces SNc and LC axonal deficits prior to somatic loss in male subjects, consistent with a dying-back pattern of degeneration, whereas female mice are resistant to chronic meth-induced degeneration.
Collapse
Affiliation(s)
| | - Steven M. Graves
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
27
|
Alshamrani M. Recent Trends in Active and Passive Immunotherapies of Alzheimer's Disease. Antibodies (Basel) 2023; 12:41. [PMID: 37366656 DOI: 10.3390/antib12020041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
In the elderly, a debilitating condition known as dementia, which is a major health concern, is caused by Alzheimer's disease (AD). Despite promising advances by researchers, there is currently no way to completely cure this devastating disease. It is illustrated by the deposition of amyloid β-peptide (Aβ) plaques that are followed by neural dysfunction and cognitive decline. Responses against AD activate an immune system that contributes to and accelerates AD pathogenesis. Potential efforts in the field of pathogenesis have prompted researchers to explore novel therapies such as active and passive vaccines against Aβ proteins (Aβ immunotherapy), intravenous immunoglobulin, and tau immunotherapy, as well as targets that include microglia and several cytokines for the treatment of AD. Aims are now underway by experts to begin immunotherapies before the clinical manifestation, which is made possible by improving the sensitivity of biomarkers used for the diagnosis of AD to have better outcome measures. This review provides an overview of approved immunotherapeutic strategies for AD and those currently being investigated in clinical trials. We examine their mechanisms of action and discuss the potential perspectives and challenges associated with immunotherapies for AD.
Collapse
Affiliation(s)
- Meshal Alshamrani
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
28
|
Martin SC, Joyce KK, Harper KM, Nikolova VD, Cohen TJ, Moy SS, Diering GH. Sleep disruption precedes forebrain synaptic Tau burden and contributes to cognitive decline in a sex-dependent manner in the P301S Tau transgenic mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.07.544101. [PMID: 37333395 PMCID: PMC10274785 DOI: 10.1101/2023.06.07.544101] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Background Sleep is an essential process that supports brain health and cognitive function in part through the modification of neuronal synapses. Sleep disruption, and impaired synaptic processes, are common features in neurodegenerative diseases, including Alzheimer's disease (AD). However, the casual role of sleep disruption in disease progression is not clear. Neurofibrillary tangles, made from hyperphosphorylated and aggregated Tau protein, form one of the major hallmark pathologies seen in AD and contribute to cognitive decline, synapse loss and neuronal death.Tau has been shown to aggregate in synapses which may impair restorative synapse processes occurring during sleep. However, it remains unclear how sleep disruption and synaptic Tau pathology interact to drive cognitive decline. It is also unclear whether the sexes show differential vulnerability to the effects of sleep loss in the context of neurodegeneration. Methods We used a piezoelectric home-cage monitoring system to measure sleep behavior in 3-11month-old transgenic hTau P301S Tauopathy model mice (PS19) and littermate controls of both sexes. Subcellular fractionation and Western blot was used to examine Tau pathology in mouse forebrain synapse fractions. To examine the role of sleep disruption in disease progression, mice were exposed to acute or chronic sleep disruption. The Morris water maze test was used to measure spatial learning and memory performance. Results PS19 mice exhibited a selective loss of sleep during the dark phase, referred to as hyperarousal, as an early symptom with an onset of 3months in females and 6months in males. At 6months, forebrain synaptic Tau burden did not correlate with sleep measures and was not affected by acute or chronic sleep disruption. Chronic sleep disruption accelerated the onset of decline of hippocampal spatial memory in PS19 males, but not females. Conclusions Dark phase hyperarousal is an early symptom in PS19 mice that precedes robust Tau aggregation. We find no evidence that sleep disruption is a direct driver of Tau pathology in the forebrain synapse. However, sleep disruption synergized with Tau pathology to accelerate the onset of cognitive decline in males. Despite the finding that hyperarousal appears earlier in females, female cognition was resilient to the effects of sleep disruption.
Collapse
|
29
|
Ehrenberg AJ, Kelberman MA, Liu KY, Dahl MJ, Weinshenker D, Falgàs N, Dutt S, Mather M, Ludwig M, Betts MJ, Winer JR, Teipel S, Weigand AJ, Eschenko O, Hämmerer D, Leiman M, Counts SE, Shine JM, Robertson IH, Levey AI, Lancini E, Son G, Schneider C, Egroo MV, Liguori C, Wang Q, Vazey EM, Rodriguez-Porcel F, Haag L, Bondi MW, Vanneste S, Freeze WM, Yi YJ, Maldinov M, Gatchel J, Satpati A, Babiloni C, Kremen WS, Howard R, Jacobs HIL, Grinberg LT. Priorities for research on neuromodulatory subcortical systems in Alzheimer's disease: Position paper from the NSS PIA of ISTAART. Alzheimers Dement 2023; 19:2182-2196. [PMID: 36642985 PMCID: PMC10182252 DOI: 10.1002/alz.12937] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/08/2022] [Accepted: 12/19/2022] [Indexed: 01/17/2023]
Abstract
The neuromodulatory subcortical system (NSS) nuclei are critical hubs for survival, hedonic tone, and homeostasis. Tau-associated NSS degeneration occurs early in Alzheimer's disease (AD) pathogenesis, long before the emergence of pathognomonic memory dysfunction and cortical lesions. Accumulating evidence supports the role of NSS dysfunction and degeneration in the behavioral and neuropsychiatric manifestations featured early in AD. Experimental studies even suggest that AD-associated NSS degeneration drives brain neuroinflammatory status and contributes to disease progression, including the exacerbation of cortical lesions. Given the important pathophysiologic and etiologic roles that involve the NSS in early AD stages, there is an urgent need to expand our understanding of the mechanisms underlying NSS vulnerability and more precisely detail the clinical progression of NSS changes in AD. Here, the NSS Professional Interest Area of the International Society to Advance Alzheimer's Research and Treatment highlights knowledge gaps about NSS within AD and provides recommendations for priorities specific to clinical research, biomarker development, modeling, and intervention. HIGHLIGHTS: Neuromodulatory nuclei degenerate in early Alzheimer's disease pathological stages. Alzheimer's pathophysiology is exacerbated by neuromodulatory nuclei degeneration. Neuromodulatory nuclei degeneration drives neuropsychiatric symptoms in dementia. Biomarkers of neuromodulatory integrity would be value-creating for dementia care. Neuromodulatory nuclei present strategic prospects for disease-modifying therapies.
Collapse
Affiliation(s)
- Alexander J Ehrenberg
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, California, USA
| | - Michael A Kelberman
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kathy Y Liu
- Division of Psychiatry, University College London, London, UK
| | - Martin J Dahl
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Neus Falgàs
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Global Brain Health Institute, University of California, San Francisco, San Francisco, California, USA
| | - Shubir Dutt
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
- Department of Psychology, University of Southern California, Los Angeles, California, USA
| | - Mara Mather
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
- Department of Psychology, University of Southern California, Los Angeles, California, USA
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Mareike Ludwig
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
| | - Matthew J Betts
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Magdeburg, Germany
| | - Joseph R Winer
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Stefan Teipel
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Rostock/Greifswald, Rostock, Germany
- Department of Psychosomatic Medicine, University Medicine Rostock, Rostock, Germany
| | - Alexandra J Weigand
- San Diego State University/University of California San Diego Joint Doctoral Program in Clinical Psychology, San Diego, California, USA
| | - Oxana Eschenko
- Department of Computational Neuroscience, Max Planck Institute for Biological Cybernetics, Tuebingen, Germany
| | - Dorothea Hämmerer
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Magdeburg, Germany
- Department of Psychology, University of Innsbruck, Innsbruck, Austria
- Institute of Cognitive Neuroscience, University College London, London, UK
| | - Marina Leiman
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Magdeburg, Germany
| | - Scott E Counts
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, Michigan, USA
- Department of Family Medicine, Michigan State University, Grand Rapids, Michigan, USA
- Michigan Alzheimer's Disease Research Center, Ann Arbor, Michigan, USA
| | - James M Shine
- Brain and Mind Center, The University of Sydney, Sydney, Australia
| | - Ian H Robertson
- Global Brain Health Institute, Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Allan I Levey
- Goizueta Alzheimer's Disease Research Center, Emory University, Atlanta, Georgia, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
- Goizueta Institute, Emory University, Atlanta, Georgia, USA
| | - Elisa Lancini
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Magdeburg, Germany
| | - Gowoon Son
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California, USA
| | - Christoph Schneider
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Maxime Van Egroo
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Faculty of Health, Medicine, and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands
| | - Claudio Liguori
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Neurology Unit, University Hospital of Rome Tor Vergata, Rome, Italy
| | - Qin Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Agusta University, Agusta, Georgia, USA
| | - Elena M Vazey
- Department of Biology, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | | | - Lena Haag
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Magdeburg, Germany
| | - Mark W Bondi
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
- Psychology Service, VA San Diego Healthcare System, San Diego, California, USA
| | - Sven Vanneste
- Global Brain Health Institute, Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- School of Psychology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Whitney M Freeze
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Neuropsychology and Psychiatry, Maastricht University, Maastricht, the Netherlands
| | - Yeo-Jin Yi
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Magdeburg, Germany
| | - Mihovil Maldinov
- Department of Psychiatry and Psychotherapy, University of Rostock, Rostock, Germany
| | - Jennifer Gatchel
- Division of Geriatric Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Abhijit Satpati
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California, USA
| | - Claudio Babiloni
- Department of Physiology and Pharmacology "V. Erspamer,", Sapienza University of Rome, Rome, Italy
- Hospital San Raffaele Cassino, Cassino, Italy
| | - William S Kremen
- Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, California, USA
| | - Robert Howard
- Division of Psychiatry, University College London, London, UK
| | - Heidi I L Jacobs
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Faculty of Health, Medicine, and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands
| | - Lea T Grinberg
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California, USA
- Global Brain Health Institute, University of California, San Francisco, San Francisco, California, USA
- Department of Pathology, University of California, San Francisco, San Francisco, California, USA
- Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
| |
Collapse
|
30
|
Torrente D, Su EJ, Schielke GP, Warnock M, Mann K, Lawrence DA. Opposing effects of β-2 and β-1 adrenergic receptor signaling on neuroinflammation and dopaminergic neuron survival in α-synuclein-mediated neurotoxicity. J Neuroinflammation 2023; 20:56. [PMID: 36864439 PMCID: PMC9983231 DOI: 10.1186/s12974-023-02748-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 02/21/2023] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND Noradrenergic neurons in the locus coeruleus (LC) are the primary source of norepinephrine (NE) in the brain and degeneration of these neurons is reported in the early stages of Parkinson's disease (PD), even prior to dopaminergic neuron degeneration in the substantia nigra (SN), which is a hallmark of PD pathology. NE depletion is generally associated with increased PD pathology in neurotoxin-based PD models. The effect of NE depletion in other models of PD-like α-synuclein-based models is largely unexplored. In PD models and in human patients, β-adrenergic receptors' (AR) signaling is associated with a reduction of neuroinflammation and PD pathology. However, the effect of NE depletion in the brain and the extent of NE and β-ARs signaling involvement in neuroinflammation, and dopaminergic neuron survival is poorly understood. METHODS Two mouse models of PD, a 6OHDA neurotoxin-based model and a human α-synuclein (hα-SYN) virus-based model of PD, were used. DSP-4 was used to deplete NE levels in the brain and its effect was confirmed by HPLC with electrochemical detection. A pharmacological approach was used to mechanistically understand the impact of DSP-4 in the hα-SYN model of PD using a norepinephrine transporter (NET) and a β-AR blocker. Epifluorescence and confocal imaging were used to study changes in microglia activation and T-cell infiltration after β1-AR and β2-AR agonist treatment in the hα-SYN virus-based model of PD. RESULTS Consistent with previous studies, we found that DSP-4 pretreatment increased dopaminergic neuron loss after 6OHDA injection. In contrast, DSP-4 pretreatment protected dopaminergic neurons after hα-SYN overexpression. DSP-4-mediated protection of dopaminergic neurons after hα-SYN overexpression was dependent on β-AR signaling since using a β-AR blocker prevented DSP-4-mediated dopaminergic neuron protection in this model of PD. Finally, we found that the β-2AR agonist, clenbuterol, reduced microglia activation, T-cell infiltration, and dopaminergic neuron degeneration, whereas xamoterol a β-1AR agonist showed increased neuroinflammation, blood brain barrier permeability (BBB), and dopaminergic neuron degeneration in the context of hα-SYN-mediated neurotoxicity. CONCLUSIONS Our data demonstrate that the effects of DSP-4 on dopaminergic neuron degeneration are model specific, and suggest that in the context of α-SYN-driven neuropathology, β2-AR specific agonists may have therapeutic benefit in PD.
Collapse
Affiliation(s)
- Daniel Torrente
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-0644, USA
| | - Enming J Su
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gerald P Schielke
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Mark Warnock
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Kris Mann
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Daniel A Lawrence
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-0644, USA.
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
31
|
Downs AM, Catavero CM, Kasten MR, McElligott ZA. Tauopathy and alcohol consumption interact to alter locus coeruleus excitatory transmission and excitability in male and female mice. Alcohol 2023; 107:97-107. [PMID: 36150608 DOI: 10.1016/j.alcohol.2022.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/22/2022] [Accepted: 08/29/2022] [Indexed: 12/23/2022]
Abstract
Alcohol use disorder is a major public health concern in the United States. Recent work has suggested a link between chronic alcohol consumption and the development of tauopathy disorders, such as Alzheimer's disease and frontotemporal dementia. However, relatively little work has investigated changes in neural circuitry involved in both tauopathy disorders and alcohol use disorder. The locus coeruleus (LC) is the major noradrenergic nucleus in the brain and is one of the earliest sites to be affected by tau lesions. The LC is also implicated in the rewarding effects of ethanol and alcohol withdrawal. In this study we assessed effects of long-term ethanol consumption and tauopathy on the physiology of LC neurons. Male and female P301S mice, a humanized transgenic mouse model of tauopathy, underwent 16 weeks of intermittent access to 20% ethanol from 3 to 7 months of age. We observed higher total alcohol consumption in female mice regardless of genotype. Male P301S mice consumed more ethanol and had a greater preference for ethanol than wild-type (WT) males. At the end of the drinking study, LC function was assessed using ex vivo whole cell electrophysiology. We found significant changes in excitatory inputs to the LC due to both ethanol and genotype. We found significantly increased excitability of the LC due to ethanol with greater effects in female P301S mice than in female WT mice. Our study identifies significant changes in the LC due to interactions between tauopathy and long-term ethanol use. These findings could have important implications regarding LC activity and changes in behavior due to both ethanol- and tauopathy-related dementia.
Collapse
Affiliation(s)
- Anthony M Downs
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Christina M Catavero
- Graduate Program in Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Michael R Kasten
- Department of Otolaryngology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Zoé A McElligott
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| |
Collapse
|
32
|
Aghakhanyan G, Galgani A, Vergallo A, Lombardo F, Martini N, Baldacci F, Tognoni G, Leo A, Guidoccio F, Siciliano G, Fornai F, Pavese N, Volterrani D, Giorgi FS. Brain metabolic correlates of Locus Coeruleus degeneration in Alzheimer's disease: a multimodal neuroimaging study. Neurobiol Aging 2023; 122:12-21. [PMID: 36463849 DOI: 10.1016/j.neurobiolaging.2022.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/11/2022]
Abstract
Locus Coeruleus (LC) degeneration occurs early in Alzheimer's disease (AD) and this could affect several brain regions innervated by LC noradrenergic axon terminals, as these bear neuroprotective effects and modulate neurovascular coupling/neuronal activity. We used LC-sensitive Magnetic Resonance imaging (MRI) sequences enabling LC integrity quantification, and [18F]Fluorodeoxyglucose (FDG) PET, to investigate the association of LC-MRI changes with brain glucose metabolism in cognitively impaired patients (30 amnesticMCI and 13 demented ones). Fifteen cognitively intact age-matched controls (HCs) were submitted only to LC-MRI for comparison with patients. Voxel-wise regression analyses of [18F]FDG images were conducted using the LC-MRI parameters signal intensity (LCCR) and LC-belonging voxels (LCVOX). Both LCCR and LCVOX were significantly lower in patients compared to HCs, and were directly associated with [18F]FDG uptake in fronto-parietal cortical areas, mainly involving the left hemisphere (p < 0.001, kE > 100). These results suggest a possible association between LC degeneration and cortical hypometabolism in degenerative cognitive impairment with a prevalent left-hemispheric vulnerability, and that LC degeneration might be linked to large-scale functional network alteration in AD pathology.
Collapse
Affiliation(s)
- Gayane Aghakhanyan
- Nuclear Medicine Unit - Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Alessandro Galgani
- Neurology Unit - Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy; Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Andrea Vergallo
- Department of Radiology, Fondazione Monasterio/CNR, Pisa, Italy
| | | | | | - Filippo Baldacci
- Neurology Unit - Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gloria Tognoni
- Neurology Unit - Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Andrea Leo
- Nuclear Medicine Unit - Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Federica Guidoccio
- Nuclear Medicine Unit - Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Gabriele Siciliano
- Nuclear Medicine Unit - Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Francesco Fornai
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Nicola Pavese
- Clinical Aging Research Unit, Newcastle University, Newcastle upon Tyne, UK; Institute of Clinical Medicine, PET Centre, Aarhus University, Aarhus, Denmark
| | - Duccio Volterrani
- Nuclear Medicine Unit - Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Filippo S Giorgi
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France.
| |
Collapse
|
33
|
Galgani A, Lombardo F, Martini N, Vergallo A, Bastiani L, Hampel H, Hlavata H, Baldacci F, Tognoni G, De Marchi D, Ghicopulos I, De Cori S, Biagioni F, Busceti CL, Ceravolo R, Bonuccelli U, Chiappino D, Siciliano G, Fornai F, Pavese N, Giorgi FS. Magnetic resonance imaging Locus Coeruleus abnormality in amnestic Mild Cognitive Impairment is associated with future progression to dementia. Eur J Neurol 2023; 30:32-46. [PMID: 36086917 PMCID: PMC10092028 DOI: 10.1111/ene.15556] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/17/2022] [Accepted: 08/31/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND PURPOSE Human neuropathological studies indicate that the pontine nucleus Locus Coeruleus (LC) undergoes significant and early degeneration in Alzheimer's disease. This line of evidence alongside experimental data suggests that the LC functional/structural decay may represent a critical factor for Alzheimer's disease pathophysiological and clinical progression. In the present prospective study, we used Magnetic Resonance Imaging (MRI) with LC-sensitive sequence (LC-MRI) to investigate in vivo the LC involvement in Alzheimer's disease progression, and whether specific LC-MRI features at baseline are associated with prognosis and cognitive performance in amnestic Mild Cognitive Impairment. METHODS LC-MRI parameters were measured at baseline by a template-based method on 3.0-T magnetic resonance images in 34 patients with Alzheimer's disease dementia, 73 patients with amnestic Mild Cognitive Impairment, and 53 cognitively intact individuals. A thorough neurological and neuropsychological assessment was performed at baseline and 2.5-year follow-up. RESULTS In subjects with Mild Cognitive Impairment who converted to dementia (n = 32), the LC intensity and number of LC-related voxels were significantly lower than in cognitively intact individuals, resembling those observed in demented patients. Such a reduction was not detected in Mild Cognitive Impairment individuals, who remained stable at follow-up. In Mild Cognitive Impairment subjects converting to dementia, LC-MRI parameter reduction was maximal in the rostral part of the left nucleus. Structural equation modeling analysis showed that LC-MRI parameters positively correlate with cognitive performance. CONCLUSIONS Our findings highlight a potential role of LC-MRI for predicting clinical progression in Mild Cognitive Impairment and support the key role of LC degeneration in the Alzheimer clinical continuum.
Collapse
Affiliation(s)
- Alessandro Galgani
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesco Lombardo
- Cardiovascular and Neuroradiological Multimodal Imaging Unit, G. Monasterio Foundation-National Research Council/Tuscany Region, Pisa, Italy
| | - Nicola Martini
- Deep Health Unit, G. Monasterio Foundation-National Research Council/Tuscany Region, Pisa, Italy
| | - Andrea Vergallo
- GRC No. 21, Alzheimer Precision Medicine, Public Hospital Network of Paris, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Luca Bastiani
- Institute of Clinical Physiology of National Research Council, Pisa, Italy
| | - Harald Hampel
- GRC No. 21, Alzheimer Precision Medicine, Public Hospital Network of Paris, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Hana Hlavata
- Cardiovascular and Neuroradiological Multimodal Imaging Unit, G. Monasterio Foundation-National Research Council/Tuscany Region, Pisa, Italy
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gloria Tognoni
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Daniele De Marchi
- Cardiovascular and Neuroradiological Multimodal Imaging Unit, G. Monasterio Foundation-National Research Council/Tuscany Region, Pisa, Italy
| | - Irene Ghicopulos
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Sara De Cori
- Cardiovascular and Neuroradiological Multimodal Imaging Unit, G. Monasterio Foundation-National Research Council/Tuscany Region, Pisa, Italy
| | - Francesca Biagioni
- Scientific Institute for Research and Health Care Neuromed, Pozzilli, Italy
| | | | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Ubaldo Bonuccelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Dante Chiappino
- Cardiovascular and Neuroradiological Multimodal Imaging Unit, G. Monasterio Foundation-National Research Council/Tuscany Region, Pisa, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesco Fornai
- Scientific Institute for Research and Health Care Neuromed, Pozzilli, Italy.,Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Nicola Pavese
- Clinical Ageing Research Unit, Newcastle University, Newcastle Upon Tyne, UK.,Institute of Clinical Medicine, Positron Emission Tomography Center, Aarhus University, Aarhus, Denmark
| | - Filippo Sean Giorgi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| |
Collapse
|
34
|
Portela Moreira I, Henriques T, Vieira-Coelho MA, Guimarães J. Dysfunction of norepinephrine and its metabolites in Alzheimer's dementia - A review with meta-analysis. Ageing Res Rev 2023; 83:101784. [PMID: 36368648 DOI: 10.1016/j.arr.2022.101784] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022]
Abstract
Some studies point locus coeruleus cell loss, the central nervous system main source of norepinephrine, to be one of the earliest neuropathological events of Alzheimer's disease (AD). However, there are conflicting reports regarding the level of norepinephrine and its metabolites (3-Methoxy-4-hydroxyphenylglycol (MHPG), 3,5-dihydroxyphenylglycine (DHPG) and 3,4 -dihydroxyphenylglycolaldehyde (DOPEGAL)) in AD patients. Uncover these alterations may be a key factor for understanding cognitive deficits and AD pathology. We review the literature that compare norepinephrine and its metabolites between AD patients and non-demented controls. A meta-analysis did not reveal significant statistical differences, but there was a trend towards a lower level of norepinephrine of AD, with almost no difference in MHPG in the cerebrospinal fluid. Regarding MHPG in plasma, DHPG and DOPEGAL we only performed a qualitative analyse due to the small or absent number of studies. These findings point to a decrease in norepinephrine, what is in line with locus coeluleus cell loss in AD. The absence of statistical difference and an equal level of MHGP could indicate a compensatory mechanism.
Collapse
Affiliation(s)
- Isabel Portela Moreira
- Clinical Neurosciences and Mental Health Department, Faculty of Medicine of Porto University, Porto, Portugal; Neurology Department, Hospital Privado de Gaia do Grupo Trofa Saúde, Vila Nova de Gaia, Portugal.
| | - Teresa Henriques
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine of Porto University, Porto, Portugal; Centre for Health Technology Services Research (CINTESIS), Faculty of Medicine of Porto University, Porto, Portugal
| | - Maria Augusta Vieira-Coelho
- Department of Biomedicine, Faculty of Medicine, Faculty of Medicine of Porto University, Porto, Portugal; Department of Psychiatry and Mental Health, University Hospital Center of São João, Porto, Portugal
| | - Joana Guimarães
- Clinical Neurosciences and Mental Health Department, Faculty of Medicine of Porto University, Porto, Portugal; Neurology Department, Centro Hospitalar Universitário de São João, Porto, Portugal
| |
Collapse
|
35
|
Iannitelli AF, Kelberman MA, Lustberg DJ, Korukonda A, McCann KE, Mulvey B, Segal A, Liles LC, Sloan SA, Dougherty JD, Weinshenker D. The Neurotoxin DSP-4 Dysregulates the Locus Coeruleus-Norepinephrine System and Recapitulates Molecular and Behavioral Aspects of Prodromal Neurodegenerative Disease. eNeuro 2023; 10:ENEURO.0483-22.2022. [PMID: 36635251 PMCID: PMC9829100 DOI: 10.1523/eneuro.0483-22.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
The noradrenergic locus coeruleus (LC) is among the earliest sites of tau and α-synuclein pathology in Alzheimer's disease (AD) and Parkinson's disease (PD), respectively. The onset of these pathologies coincides with loss of noradrenergic fibers in LC target regions and the emergence of prodromal symptoms including sleep disturbances and anxiety. Paradoxically, these prodromal symptoms are indicative of a noradrenergic hyperactivity phenotype, rather than the predicted loss of norepinephrine (NE) transmission following LC damage, suggesting the engagement of complex compensatory mechanisms. Because current therapeutic efforts are targeting early disease, interest in the LC has grown, and it is critical to identify the links between pathology and dysfunction. We employed the LC-specific neurotoxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4), which preferentially damages LC axons, to model early changes in the LC-NE system pertinent to AD and PD in male and female mice. DSP-4 (two doses of 50 mg/kg, one week apart) induced LC axon degeneration, triggered neuroinflammation and oxidative stress, and reduced tissue NE levels. There was no LC cell death or changes to LC firing, but transcriptomics revealed reduced expression of genes that define noradrenergic identity and other changes relevant to neurodegenerative disease. Despite the dramatic loss of LC fibers, NE turnover and signaling were elevated in terminal regions and were associated with anxiogenic phenotypes in multiple behavioral tests. These results represent a comprehensive analysis of how the LC-NE system responds to axon/terminal damage reminiscent of early AD and PD at the molecular, cellular, systems, and behavioral levels, and provides potential mechanisms underlying prodromal neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Alexa F Iannitelli
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - Michael A Kelberman
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - Daniel J Lustberg
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - Anu Korukonda
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - Katharine E McCann
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - Bernard Mulvey
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
| | - Arielle Segal
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - L Cameron Liles
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - Steven A Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - Joseph D Dougherty
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
36
|
Mercan D, Heneka MT. The Contribution of the Locus Coeruleus-Noradrenaline System Degeneration during the Progression of Alzheimer's Disease. BIOLOGY 2022; 11:1822. [PMID: 36552331 PMCID: PMC9775634 DOI: 10.3390/biology11121822] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD), which is characterized by extracellular accumulation of amyloid-beta peptide and intracellular aggregation of hyperphosphorylated tau, is the most common form of dementia. Memory loss, cognitive decline and disorientation are the ultimate consequences of neuronal death, synapse loss and neuroinflammation in AD. In general, there are many brain regions affected but neuronal loss in the locus coeruleus (LC) is one of the earliest indicators of neurodegeneration in AD. Since the LC is the main source of noradrenaline (NA) in the brain, degeneration of the LC in AD leads to decreased NA levels, causing increased neuroinflammation, enhanced amyloid and tau burden, decreased phagocytosis and impairment in cognition and long-term synaptic plasticity. In this review, we summarized current findings on the locus coeruleus-noradrenaline system and consequences of its dysfunction which is now recognized as an important contributor to AD progression.
Collapse
Affiliation(s)
- Dilek Mercan
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Michael Thomas Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
37
|
Kapri D, Vadodaria KC, Rommelfanger KS, Ogbonmwan YE, Liles LC, Fernandes-Thomas KA, Salvi SS, Husain BF, Weinshenker D, Vaidya VA. Genetic loss of norepinephrine does not alter adult hippocampal neurogenesis in dopamine beta-hydroxylase deficient mice. IBRO Neurosci Rep 2022; 13:420-425. [PMID: 36386600 PMCID: PMC9643407 DOI: 10.1016/j.ibneur.2022.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/25/2022] [Indexed: 11/07/2022] Open
Abstract
Norepinephrine (NE), and specific adrenoceptors, have been reported to influence distinct aspects of adult hippocampal neurogenesis, including latent stem cell activation, progenitor proliferation, and differentiation. These findings are predominantly based on the use of pharmacological approaches in both in vitro and in vivo systems. Here, we sought to assess the consequences of genetic ablation of NE on adult hippocampal neurogenesis, by examining dopamine β hydroxylase knockout (Dbh -/-) mice, which lack NE from birth. We find that Dbh -/- mice exhibit no difference in adult hippocampal progenitor proliferation and survival. Further, the number of immature newborn neurons, labeled using stage-specific developmental markers within the hippocampal neurogenic niche, was also unaltered in Dbh -/- mice. In contrast, the noradrenergic neurotoxin DSP-4, which had previously been shown to reduce adult hippocampal neurogenesis in rats, also resulted in a decline in hippocampal progenitor proliferation in C57/Bl6N mice. These findings indicate that pharmacological lesioning of noradrenergic afferents in adulthood, but not the complete genetic loss of NE from birth, impairs adult hippocampal neurogenesis in mice.
Collapse
Affiliation(s)
- Darshana Kapri
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Krishna C. Vadodaria
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | | | | | - L. Cameron Liles
- Department of Human Genetics, Emory University, Atlanta, GA 30322, USA
| | | | - Sonali S. Salvi
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Basma F.A. Husain
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - David Weinshenker
- Department of Human Genetics, Emory University, Atlanta, GA 30322, USA
| | - Vidita A. Vaidya
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| |
Collapse
|
38
|
Elman JA, Puckett OK, Hagler DJ, Pearce RC, Fennema-Notestine C, Hatton SN, Lyons MJ, McEvoy LK, Panizzon MS, Reas ET, Dale AM, Franz CE, Kremen WS. Associations between MRI-assessed locus coeruleus integrity and cortical gray matter microstructure. Cereb Cortex 2022; 32:4191-4203. [PMID: 34969072 PMCID: PMC9528780 DOI: 10.1093/cercor/bhab475] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/17/2021] [Accepted: 11/22/2021] [Indexed: 01/27/2023] Open
Abstract
The locus coeruleus (LC) is one of the earliest sites of tau pathology, making it a key structure in early Alzheimer's disease (AD) progression. As the primary source of norepinephrine for the brain, reduced LC integrity may have negative consequences for brain health, yet macrostructural brain measures (e.g. cortical thickness) may not be sensitive to early stages of neurodegeneration. We therefore examined whether LC integrity was associated with differences in cortical gray matter microstructure among 435 men (mean age = 67.5; range = 62-71.7). LC structural integrity was indexed by contrast-to-noise ratio (LCCNR) from a neuromelanin-sensitive MRI scan. Restriction spectrum imaging (RSI), an advanced multi-shell diffusion technique, was used to characterize cortical microstructure, modeling total diffusion in restricted, hindered, and free water compartments. Higher LCCNR (greater integrity) was associated with higher hindered and lower free water diffusion in multiple cortical regions. In contrast, no associations between LCCNR and cortical thickness survived correction. Results suggest lower LC integrity is associated with patterns of cortical microstructure that may reflect a reduction in cytoarchitectural barriers due to broader neurodegenerative processes. These findings highlight the potential utility for LC imaging and advanced diffusion measures of cortical microstructure in assessing brain health and early identification of neurodegenerative processes.
Collapse
Affiliation(s)
- Jeremy A Elman
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
| | - Olivia K Puckett
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
| | - Donald J Hagler
- Department of Radiology, University of California San Diego, La Jolla, CA 92093, USA
| | - Rahul C Pearce
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
| | - Christine Fennema-Notestine
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- Department of Radiology, University of California San Diego, La Jolla, CA 92093, USA
| | - Sean N Hatton
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Michael J Lyons
- Department of Psychological and Brain Sciences, Boston University, Boston, MA 02215, USA
| | - Linda K McEvoy
- Department of Radiology, University of California San Diego, La Jolla, CA 92093, USA
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, CA 92093, USA
| | - Matthew S Panizzon
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
| | - Emilie T Reas
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Anders M Dale
- Department of Radiology, University of California San Diego, La Jolla, CA 92093, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Carol E Franz
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
| | - William S Kremen
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
- Center of Excellence for Stress and Mental Health, VA San Diego Health Care System, La Jolla, CA 92161, USA
| |
Collapse
|
39
|
Alfaro‐Ruiz R, Aguado C, Martín‐Belmonte A, Moreno‐Martínez AE, Merchán‐Rubira J, Hernández F, Ávila J, Fukazawa Y, Luján R. Different modes of synaptic and extrasynaptic NMDA receptor alteration in the hippocampus of P301S tau transgenic mice. Brain Pathol 2022; 33:e13115. [PMID: 36058615 PMCID: PMC9836375 DOI: 10.1111/bpa.13115] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/12/2022] [Indexed: 01/21/2023] Open
Abstract
N-methyl-d-aspartate receptors (NMDARs) are pivotal players in the synaptic transmission and synaptic plasticity underlying learning and memory. Accordingly, dysfunction of NMDARs has been implicated in the pathophysiology of Alzheimer disease (AD). Here, we used histoblot and sodium dodecylsulphate-digested freeze-fracture replica labelling (SDS-FRL) techniques to investigate the expression and subcellular localisation of GluN1, the obligatory subunit of NMDARs, in the hippocampus of P301S mice. Histoblots showed that GluN1 expression was significantly reduced in the hippocampus of P301S mice in a laminar-specific manner at 10 months of age but was unaltered at 3 months. Using the SDS-FRL technique, excitatory synapses and extrasynaptic sites on spines of pyramidal cells and interneuron dendrites were analysed throughout all dendritic layers in the CA1 field. Our ultrastructural approach revealed a high density of GluN1 in synaptic sites and a substantially lower density at extrasynaptic sites. Labelling density for GluN1 in excitatory synapses established on spines was significantly reduced in P301S mice, compared with age-matched wild-type mice, in the stratum oriens (so), stratum radiatum (sr) and stratum lacunosum-moleculare (slm). Density for synaptic GluN1 on interneuron dendrites was significantly reduced in P301S mice in the so and sr but unaltered in the slm. Labelling density for GluN1 at extrasynaptic sites showed no significant differences in pyramidal cells, and only increased density in the interneuron dendrites of the sr. This differential alteration of synaptic versus extrasynaptic NMDARs supports the notion that the progressive accumulation of phospho-tau is associated with changes in NMDARs, in the absence of amyloid-β pathology, and may be involved in the mechanisms causing abnormal network activity of the hippocampal circuit.
Collapse
Affiliation(s)
- Rocío Alfaro‐Ruiz
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de MedicinaUniversidad Castilla‐La Mancha, Campus BiosanitarioAlbaceteSpain
| | - Carolina Aguado
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de MedicinaUniversidad Castilla‐La Mancha, Campus BiosanitarioAlbaceteSpain
| | - Alejandro Martín‐Belmonte
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de MedicinaUniversidad Castilla‐La Mancha, Campus BiosanitarioAlbaceteSpain,Present address:
Pharmacology Unit, Department of Pathology and Experimental TherapeuticsFaculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona08907 L'Hospitalet de LlobregatSpain
| | - Ana Esther Moreno‐Martínez
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de MedicinaUniversidad Castilla‐La Mancha, Campus BiosanitarioAlbaceteSpain
| | | | - Félix Hernández
- Centro de Biología Molecular Severo OchoaCSIC‐UAMMadridSpain,Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasISCIIIMadridSpain
| | - Jesús Ávila
- Centro de Biología Molecular Severo OchoaCSIC‐UAMMadridSpain,Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasISCIIIMadridSpain
| | - Yugo Fukazawa
- Division of Brain Structure and Function, Faculty of Medical ScienceUniversity of FukuiFukuiJapan,Life Science Innovation CenterUniversity of FukuiFukuiJapan
| | - Rafael Luján
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de MedicinaUniversidad Castilla‐La Mancha, Campus BiosanitarioAlbaceteSpain
| |
Collapse
|
40
|
Kelberman MA, Weinshenker D. A novel link between locus coeruleus activity and amyloid-related cognitive decline. Trends Neurosci 2022; 45:651-653. [PMID: 35659415 PMCID: PMC10216710 DOI: 10.1016/j.tins.2022.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 01/28/2023]
Abstract
Recent work from Prokopiou, Engels-Domínguez et al. assessed locus coeruleus (LC) activity and its functional connectivity (FC) to forebrain regions during a novelty task in cognitively unimpaired adult individuals with varying degrees of amyloid deposition. Novelty increased LC activity and LC FC, but lower responses on these measures were associated with steeper cognitive decline in amyloid-positive individuals.
Collapse
Affiliation(s)
- Michael A Kelberman
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
41
|
Flores-Aguilar L, Hall H, Orciani C, Foret MK, Kovecses O, Ducatenzeiler A, Cuello AC. Early loss of locus coeruleus innervation promotes cognitive and neuropathological changes before amyloid plaque deposition in a transgenic rat model of Alzheimer's disease. Neuropathol Appl Neurobiol 2022; 48:e12835. [PMID: 35822518 DOI: 10.1111/nan.12835] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 05/31/2022] [Accepted: 07/06/2022] [Indexed: 11/29/2022]
Abstract
AIMS The locus coeruleus (LC) is the main source of noradrenaline (NA) in the mammalian brain and has been found to degenerate during the initial stages of Alzheimer's disease (AD). Recent studies indicate that at late stages of the amyloid pathology, LC-pathological alterations accelerate AD-like pathology progression by interfering with the neuromodulatory and anti-inflammatory properties of NA. However, the impact of LC degeneration at the earliest stages of amyloidosis on the AD-like pathology is not well understood. METHODS The LC was lesioned in wild-type (wt) and McGill-R-Thy1-APP transgenic rats (APP tg) by administering N-(2-chloroethyl)-N-ethyl-bromo-benzylamine (DSP4) before amyloid plaque deposition. Cognitive deficits and AD-like neuropathological changes were measured after the LC lesion. RESULTS Four months post-treatment, rats displayed a decrease in brain noradrenergic innervation. The LC lesion in APP tg-treated rats enhanced cognitive deficits and decreased hippocampal cholinergic innervation and neurotrophin expression. In addition, the APP tg-treated rats displayed an increased microglial and astroglial cell number in close vicinity to hippocampal amyloid-beta burdened neurons. The recruited microglia showed cellular alterations indicative of an intermediate activation state. CONCLUSIONS Our results indicate that early LC demise aggravates the early neuroinflammatory process, cognitive impairments, cholinergic deficits and neurotrophin deregulation at the earliest stages of the human-like brain amyloidosis.
Collapse
Affiliation(s)
- Lisi Flores-Aguilar
- Department of Anatomy and Cell Biology, McGill University, Montreal, Canada.,Current affiliation: Department of Pathology and Laboratory Medicine, University of California, Irvine, United States of America
| | - Hélène Hall
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Chiara Orciani
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
| | - Morgan K Foret
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Olivia Kovecses
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | | | - A Claudio Cuello
- Department of Anatomy and Cell Biology, McGill University, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Visiting Professor, Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
42
|
Gutiérrez IL, Dello Russo C, Novellino F, Caso JR, García-Bueno B, Leza JC, Madrigal JLM. Noradrenaline in Alzheimer's Disease: A New Potential Therapeutic Target. Int J Mol Sci 2022; 23:ijms23116143. [PMID: 35682822 PMCID: PMC9181823 DOI: 10.3390/ijms23116143] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 12/13/2022] Open
Abstract
A growing body of evidence demonstrates the important role of the noradrenergic system in the pathogenesis of many neurodegenerative processes, especially Alzheimer’s disease, due to its ability to control glial activation and chemokine production resulting in anti-inflammatory and neuroprotective effects. Noradrenaline involvement in this disease was first proposed after finding deficits of noradrenergic neurons in the locus coeruleus from Alzheimer’s disease patients. Based on this, it has been hypothesized that the early loss of noradrenergic projections and the subsequent reduction of noradrenaline brain levels contribute to cognitive dysfunctions and the progression of neurodegeneration. Several studies have focused on analyzing the role of noradrenaline in the development and progression of Alzheimer’s disease. In this review we summarize some of the most relevant data describing the alterations of the noradrenergic system normally occurring in Alzheimer’s disease as well as experimental studies in which noradrenaline concentration was modified in order to further analyze how these alterations affect the behavior and viability of different nervous cells. The combination of the different studies here presented suggests that the maintenance of adequate noradrenaline levels in the central nervous system constitutes a key factor of the endogenous defense systems that help prevent or delay the development of Alzheimer’s disease. For this reason, the use of noradrenaline modulating drugs is proposed as an interesting alternative therapeutic option for Alzheimer’s disease.
Collapse
Affiliation(s)
- Irene L. Gutiérrez
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
| | - Cinzia Dello Russo
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology (ISMIB), University of Liverpool, Liverpool L69 3GL, UK
| | - Fabiana Novellino
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council, 88100 Catanzaro, Italy
| | - Javier R. Caso
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
| | - Borja García-Bueno
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
| | - Juan C. Leza
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
| | - José L. M. Madrigal
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
- Correspondence: ; Tel.: +34-91-394-1463
| |
Collapse
|
43
|
Zong B, Yu F, Zhang X, Zhao W, Sun P, Li S, Li L. Understanding How Physical Exercise Improves Alzheimer’s Disease: Cholinergic and Monoaminergic Systems. Front Aging Neurosci 2022; 14:869507. [PMID: 35663578 PMCID: PMC9158463 DOI: 10.3389/fnagi.2022.869507] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/14/2022] [Indexed: 01/11/2023] Open
Abstract
Alzheimer’s disease (AD) is an age-related neurodegenerative disorder, characterized by the accumulation of proteinaceous aggregates and neurofibrillary lesions composed of β-amyloid (Aβ) peptide and hyperphosphorylated microtubule-associated protein tau, respectively. It has long been known that dysregulation of cholinergic and monoaminergic (i.e., dopaminergic, serotoninergic, and noradrenergic) systems is involved in the pathogenesis of AD. Abnormalities in neuronal activity, neurotransmitter signaling input, and receptor function exaggerate Aβ deposition and tau hyperphosphorylation. Maintenance of normal neurotransmission is essential to halt AD progression. Most neurotransmitters and neurotransmitter-related drugs modulate the pathology of AD and improve cognitive function through G protein-coupled receptors (GPCRs). Exercise therapies provide an important alternative or adjunctive intervention for AD. Cumulative evidence indicates that exercise can prevent multiple pathological features found in AD and improve cognitive function through delaying the degeneration of cholinergic and monoaminergic neurons; increasing levels of acetylcholine, norepinephrine, serotonin, and dopamine; and modulating the activity of certain neurotransmitter-related GPCRs. Emerging insights into the mechanistic links among exercise, the neurotransmitter system, and AD highlight the potential of this intervention as a therapeutic approach for AD.
Collapse
Affiliation(s)
- Boyi Zong
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
- College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Fengzhi Yu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
- College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Xiaoyou Zhang
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
- College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Wenrui Zhao
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
- College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Peng Sun
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
- College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Shichang Li
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
- College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Lin Li
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
- College of Physical Education and Health, East China Normal University, Shanghai, China
- *Correspondence: Lin Li,
| |
Collapse
|
44
|
Shiu FH, Wong JC, Yamamoto T, Lala T, Purcell RH, Owino S, Zhu D, Van Meir EG, Hall RA, Escayg A. Mice lacking full length Adgrb1 (Bai1) exhibit social deficits, increased seizure susceptibility, and altered brain development. Exp Neurol 2022; 351:113994. [PMID: 35114205 PMCID: PMC9817291 DOI: 10.1016/j.expneurol.2022.113994] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 12/20/2021] [Accepted: 01/24/2022] [Indexed: 01/11/2023]
Abstract
The adhesion G protein-coupled receptor BAI1/ADGRB1 plays an important role in suppressing angiogenesis, mediating phagocytosis, and acting as a brain tumor suppressor. BAI1 is also a critical regulator of dendritic spine and excitatory synapse development and interacts with several autism-relevant proteins. However, little is known about the relationship between altered BAI1 function and clinically relevant phenotypes. Therefore, we studied the effect of reduced expression of full length Bai1 on behavior, seizure susceptibility, and brain morphology in Adgrb1 mutant mice. We compared homozygous (Adgrb1-/-), heterozygous (Adgrb1+/-), and wild-type (WT) littermates using a battery of tests to assess social behavior, anxiety, repetitive behavior, locomotor function, and seizure susceptibility. We found that Adgrb1-/- mice showed significant social behavior deficits and increased vulnerability to seizures. Adgrb1-/- mice also showed delayed growth and reduced brain weight. Furthermore, reduced neuron density and increased apoptosis during brain development were observed in the hippocampus of Adgrb1-/- mice, while levels of astrogliosis and microgliosis were comparable to WT littermates. These results show that reduced levels of full length Bai1 is associated with a broader range of clinically relevant phenotypes than previously reported.
Collapse
Affiliation(s)
- Fu Hung Shiu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA; Neuroscience Graduate Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, USA
| | - Jennifer C Wong
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Takahiro Yamamoto
- Department of Neurosurgery, School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Trisha Lala
- Neuroscience Graduate Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, USA; Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ryan H Purcell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sharon Owino
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Dan Zhu
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Erwin G Van Meir
- Department of Neurosurgery, School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Randy A Hall
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Andrew Escayg
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
45
|
Malatt C, Tagliati M. The role of the locus coeruleus/norepinephrine system in the pathogenesis of neurodegenerative disorders: An update. Curr Opin Neurol 2022; 35:220-229. [PMID: 35175974 DOI: 10.1097/wco.0000000000001042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The aim of this review was to provide an update on current and emerging knowledge of the neuropathological processes affecting the locus coeruleus/norepinephrine (LC/NE) system, their effect on Alzheimer's disease and Parkinson's disease symptomatology, including efforts to translate these notions into therapeutic actions targeting the noradrenergic system. RECENT FINDINGS Over the past 2 years, work from multiple groups has contributed to support an early role of locus coeruleus degeneration and/or hyperactivation in the neurodegenerative process, including a trigger of neuroinflammation. Imaging advances are allowing the quantification of locus coeruleus structural features in vivo, which is critical in the early stages of disease. Nonmotor and noncognitive symptoms, often secondary to the involvement of the LC/NE system, are becoming more important in the definition of these diseases and their treatment. SUMMARY The diverse symptomatology of Parkinson's disease and Alzheimer's disease, which is not limited to cardinal motor and cognitive abnormalities, strongly suggests a multisystem neurodegenerative process. In this context, it is increasingly clear how the LC/NE system plays a key role in the initiation and maintenance of the neurodegenerative process.
Collapse
Affiliation(s)
- Camille Malatt
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | | |
Collapse
|
46
|
Dahl MJ, Mather M, Werkle-Bergner M, Kennedy BL, Guzman S, Hurth K, Miller CA, Qiao Y, Shi Y, Chui HC, Ringman JM. Locus coeruleus integrity is related to tau burden and memory loss in autosomal-dominant Alzheimer's disease. Neurobiol Aging 2022; 112:39-54. [PMID: 35045380 PMCID: PMC8976827 DOI: 10.1016/j.neurobiolaging.2021.11.006] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 11/17/2021] [Accepted: 11/26/2021] [Indexed: 12/14/2022]
Abstract
Abnormally phosphorylated tau, an indicator of Alzheimer's disease, accumulates in the first decades of life in the locus coeruleus (LC), the brain's main noradrenaline supply. However, technical challenges in in-vivo assessments have impeded research into the role of the LC in Alzheimer's disease. We studied participants with or known to be at-risk for mutations in genes causing autosomal-dominant Alzheimer's disease (ADAD) with early onset, providing a unique window into the pathogenesis of Alzheimer's largely disentangled from age-related factors. Using high-resolution MRI and tau PET, we found lower rostral LC integrity in symptomatic participants. LC integrity was associated with individual differences in tau burden and memory decline. Post-mortem analyses in a separate set of carriers of the same mutation confirmed substantial neuronal loss in the LC. Our findings link LC degeneration to tau burden and memory in Alzheimer's, and highlight a role of the noradrenergic system in this neurodegenerative disease.
Collapse
Affiliation(s)
- Martin J Dahl
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany; Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
| | - Mara Mather
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Markus Werkle-Bergner
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany
| | - Briana L Kennedy
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA; School of Psychological Science, University of Western Australia, Perth, Australia
| | - Samuel Guzman
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kyle Hurth
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Carol A Miller
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yuchuan Qiao
- Laboratory of Neuro Imaging (LONI), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yonggang Shi
- Laboratory of Neuro Imaging (LONI), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Helena C Chui
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - John M Ringman
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
47
|
Kang SS, Meng L, Zhang X, Wu Z, Mancieri A, Xie B, Liu X, Weinshenker D, Peng J, Zhang Z, Ye K. Tau modification by the norepinephrine metabolite DOPEGAL stimulates its pathology and propagation. Nat Struct Mol Biol 2022; 29:292-305. [PMID: 35332321 PMCID: PMC9018606 DOI: 10.1038/s41594-022-00745-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 02/09/2022] [Indexed: 12/12/2022]
Abstract
The noradrenergic locus ceruleus (LC) is the first site of detectable tau pathology in Alzheimer's disease (AD), but the mechanisms underlying the selective vulnerability of the LC in AD have not been completely identified. In the present study, we show that DOPEGAL, a monoamine oxidase A (MAO-A) metabolite of norepinephrine (NE), reacts directly with the primary amine on the Lys353 residue of tau to stimulate its aggregation and facilitate its propagation. Inhibition of MAO-A or mutation of the Lys353 residue to arginine (Lys353Arg) decreases tau Lys353-DOPEGAL levels and diminishes tau pathology spreading. Wild-type tau preformed fibrils (PFFs) trigger Lys353-DOPEGAL formation, tau pathology propagation and cognitive impairment in MAPT transgenic mice, all of which are attenuated with PFFs made from the Lys353Arg mutant. Thus, the selective vulnerability of LC neurons in AD may be explained, in part, by NE oxidation via MAO-A into DOPEGAL, which covalently modifies tau and accelerates its aggregation, toxicity and propagation.
Collapse
Affiliation(s)
- Seong Su Kang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Lanxia Meng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xingyu Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhiping Wu
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ariana Mancieri
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Boer Xie
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA.
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Shenzhen, China.
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Shenzhen, China.
| |
Collapse
|
48
|
Ciampa CJ, Parent JH, Harrison TM, Fain RM, Betts MJ, Maass A, Winer JR, Baker SL, Janabi M, Furman DJ, D'Esposito M, Jagust WJ, Berry AS. Associations among locus coeruleus catecholamines, tau pathology, and memory in aging. Neuropsychopharmacology 2022; 47:1106-1113. [PMID: 35034099 PMCID: PMC8938463 DOI: 10.1038/s41386-022-01269-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/16/2021] [Accepted: 01/04/2022] [Indexed: 12/20/2022]
Abstract
The locus coeruleus (LC) is the brain's major source of the neuromodulator norepinephrine, and is also profoundly vulnerable to the development of Alzheimer's disease (AD)-related tau pathology. Norepinephrine plays a role in neuroprotective functions that may reduce AD progression, and also underlies optimal memory performance. Successful maintenance of LC neurochemical function represents a candidate mechanism of protection against the propagation of AD-related pathology and may facilitate the preservation of memory performance despite pathology. Using [18F]Fluoro-m-tyrosine ([18F]FMT) PET imaging to measure catecholamine synthesis capacity in LC regions of interest, we examined relationships among LC neurochemical function, AD-related pathology, and memory performance in cognitively normal older adults (n = 49). Participants underwent [11C]Pittsburgh compound B and [18F]Flortaucipir PET to quantify β-amyloid (n = 49) and tau burden (n = 42) respectively. In individuals with substantial β-amyloid, higher LC [18F]FMT net tracer influx (Kivis) was associated with lower temporal tau. Longitudinal tau-PET analyses in a subset of our sample (n = 30) support these findings to reveal reduced temporal tau accumulation in the context of higher LC [18F]FMT Kivis. Higher LC catecholamine synthesis capacity was positively correlated with self-reported cognitive engagement and physical activity across the lifespan, established predictors of successful aging measured with the Lifetime Experiences Questionnaire. LC catecholamine synthesis capacity moderated tau's negative effect on memory, such that higher LC catecholamine synthesis capacity was associated with better-than-expected memory performance given an individual's tau burden. These PET findings provide insight into the neurochemical mechanisms of AD vulnerability and cognitive resilience in the living human brain.
Collapse
Affiliation(s)
- Claire J Ciampa
- Department of Psychology, Brandeis University, Waltham, MA, 02453, USA
| | - Jourdan H Parent
- Department of Psychology, Brandeis University, Waltham, MA, 02453, USA
| | - Theresa M Harrison
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Rebekah M Fain
- Department of Psychology, Brandeis University, Waltham, MA, 02453, USA
| | - Matthew J Betts
- Institute of Cognitive Neurology and Dementia Research, Otto von Guericke University, Magdeburg, 39106, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen, Magdeburg, 39120, Germany
- Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
| | - Anne Maass
- Deutsches Zentrum für Neurodegenerative Erkrankungen, Magdeburg, 39120, Germany
| | - Joseph R Winer
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Suzanne L Baker
- Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Mustafa Janabi
- Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Daniella J Furman
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, 94720, USA
- University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Mark D'Esposito
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, 94720, USA
- Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Anne S Berry
- Department of Psychology, Brandeis University, Waltham, MA, 02453, USA.
- Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA.
| |
Collapse
|
49
|
Giliberto L. Editorial: Degenerative and cognitive diseases. Curr Opin Neurol 2022; 35:208-211. [PMID: 35232933 DOI: 10.1097/wco.0000000000001037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Luca Giliberto
- Litwin-Zucker Center for the Study of Alzheimer's Diseases and Memory Disorders, Feinstein Institutes for Medical Research and Institute for Neurology and Neurosurgery, Northwell Health System, Manhasset, New York, USA
| |
Collapse
|
50
|
Kelberman MA, Anderson CR, Chlan E, Rorabaugh JM, McCann KE, Weinshenker D. Consequences of Hyperphosphorylated Tau in the Locus Coeruleus on Behavior and Cognition in a Rat Model of Alzheimer's Disease. J Alzheimers Dis 2022; 86:1037-1059. [PMID: 35147547 PMCID: PMC9007891 DOI: 10.3233/jad-215546] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The locus coeruleus (LC) is one of the earliest brain regions to accumulate hyperphosphorylated tau, but a lack of animal models that recapitulate this pathology has hampered our understanding of its contributions to Alzheimer's disease (AD) pathophysiology. OBJECTIVE We previously reported that TgF344-AD rats, which overexpress mutant human amyloid precursor protein and presenilin-1, accumulate early endogenous hyperphosphorylated tau in the LC. Here, we used TgF344-AD rats and a wild-type (WT) human tau virus to interrogate the effects of endogenous hyperphosphorylated rat tau and human tau in the LC on AD-related neuropathology and behavior. METHODS Two-month-old TgF344-AD and WT rats received bilateral LC infusions of full-length WT human tau or mCherry control virus driven by the noradrenergic-specific PRSx8 promoter. Rats were subsequently assessed at 6 and 12 months for arousal (sleep latency), anxiety-like behavior (open field, elevated plus maze, novelty-suppressed feeding), passive coping (forced swim task), and learning and memory (Morris water maze and fear conditioning). Hippocampal microglia, astrocyte, and AD pathology were evaluated using immunohistochemistry. RESULTS In general, the effects of age were more pronounced than genotype or treatment; older rats displayed greater hippocampal pathology, took longer to fall asleep, had reduced locomotor activity, floated more, and had impaired cognition compared to younger animals. TgF344-AD rats showed increased anxiety-like behavior and impaired learning and memory. The tau virus had negligible influence on most measures. CONCLUSION Effects of hyperphosphorylated tau on AD-like neuropathology and behavioral symptoms were subtle. Further investigation of different forms of tau is warranted.
Collapse
Affiliation(s)
- Michael A. Kelberman
- Department of Human Genetics, Emory University, Atlanta, GA 30322
- Neuroscience Program, Laney Graduate School, Emory University, Atlanta, GA 30322
| | | | - Eli Chlan
- Neuroscience Program, Laney Graduate School, Emory University, Atlanta, GA 30322
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332
| | | | | | | |
Collapse
|