1
|
Shembel AC, Johnson AM, Ciucci MR, Lunaris CL, Morrison RA, Rudisch DM. The role of ultrasonic vocalizations in rat laryngological investigations. Physiol Behav 2025; 294:114887. [PMID: 40118131 DOI: 10.1016/j.physbeh.2025.114887] [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: 01/02/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 03/23/2025]
Abstract
Rat ultrasonic vocalizations (USVs) have traditionally been used in psychosocial and psychobiological studies to understand emotion, social behavior, cognition, and associative learning. However, recent studies have expanded the goal of USVs to include the study of the laryngeal system and the effects of disease processes on vocal sensorimotor control. Without the foundational understanding of the goals of this area of laryngological research, fundamental differences in study objectives between psychobehavioral and laryngological studies can easily be missed, leading to misconceptions and misinterpretations of the role USVs play in laryngology-focused studies. Standardization of terminology and methods are also needed to improve communication, enhance study replicability, and prevent ambiguity that can lead to misinterpretations of study objectives and findings in this line of research. The primary objective is to describe the role of USVs in studies of laryngeal anatomy and physiology, with a focus on their connections to the neuromuscular and neurological aspects of the laryngeal system, particularly in relation to vocal sensorimotor control and voice disorders. It is intended for novice investigators interested in laryngology-specific USV research. Researchers experienced in USV studies within the context of the larynx and vocal sensorimotor control first outline the development and refinement of various USV elicitation methods. They provide insights into how these approaches have been tested across different studies and laboratories. Finally, they advocate for standardizing terminology and methodologies to enhance study replicability, reduce ambiguity, and foster collaboration across research groups.
Collapse
Affiliation(s)
- Adrianna C Shembel
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA; Department of Otolaryngology-Head and Neck, Voice Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Aaron M Johnson
- Department of Otolaryngology-Head and Neck Surgery, NYU Voice Center, NYU Langone Health, New York, NY, USA
| | - Michelle R Ciucci
- Department of Surgery, Division of Otolaryngology-Head & Neck Surgery, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, USA; Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Robert A Morrison
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA; Department of Otolaryngology-Head and Neck, Voice Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Denis Michael Rudisch
- Department of Surgery, Division of Otolaryngology-Head & Neck Surgery, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, USA; Institute for Clinical and Translational Research, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
2
|
Černotová D, Hrůzová K, Touš J, Janča R, Stuchlík A, Levčík D, Svoboda J. Early social deficits in TgF344-AD rats are accompanied by sex-specific parvalbumin-positive interneuron reduction and altered brain oscillations in the hippocampal CA2. Neurobiol Dis 2025; 208:106875. [PMID: 40097074 DOI: 10.1016/j.nbd.2025.106875] [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: 12/05/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/19/2025] Open
Abstract
Social withdrawal and deficits in social cognition are hallmarks of Alzheimer's disease (AD). While early deficits in social behavior and memory have been documented in mouse AD models, they remain understudied in rat models. Early-stage AD is accompanied by dysfunction of parvalbumin-positive (PV+) interneurons, implicating their potential connection to early symptoms. In this study, we employed a 5-trial social memory task to investigate early deficits in social cognition in 6-month-old TgF344-AD male and female rats. We counted the number of PV+ interneurons and recorded local field potentials during social interactions in the hippocampal CA2 - a region critical for social information processing. Our results show decreased social interest and novelty preference in TgF344-AD male and female rats. However, reduced PV+ interneuron numbers were observed only in female rats and specific to the CA2 area. The electrophysiological recordings revealed reduced theta-gamma phase-amplitude coupling in the CA2 during direct social interactions. We conclude that deficits in social cognition accompany early-stage AD in TgF344-AD rats and are potentially linked to PV+ interneuron and brain oscillatory dysfunction in the CA2 region of the hippocampus.
Collapse
Affiliation(s)
- Daniela Černotová
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 142 00, Czech Republic; Third Faculty of Medicine, Charles University, Ruska 87, Prague 100 00, Czech Republic
| | - Karolína Hrůzová
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 142 00, Czech Republic; Third Faculty of Medicine, Charles University, Ruska 87, Prague 100 00, Czech Republic
| | - Jan Touš
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 142 00, Czech Republic; Faculty of Electrical Engineering, Czech Technical University in Prague, Technicka 2, Prague 160 00, Czech Republic
| | - Radek Janča
- Faculty of Electrical Engineering, Czech Technical University in Prague, Technicka 2, Prague 160 00, Czech Republic
| | - Aleš Stuchlík
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 142 00, Czech Republic
| | - David Levčík
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 142 00, Czech Republic.
| | - Jan Svoboda
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 142 00, Czech Republic
| |
Collapse
|
3
|
Marsland P, Vore AS, Lutzke A, Gano A, Fischer A, Trapp S, Savage LM, Deak T. Sex-specific effects of chronic alcohol consumption across the lifespan in the transgenic Alzheimer's Disease (TgF344-AD) rat model. Brain Behav Immun 2025; 128:192-207. [PMID: 40187669 DOI: 10.1016/j.bbi.2025.03.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 03/25/2025] [Accepted: 03/29/2025] [Indexed: 04/07/2025] Open
Abstract
Alcohol consumption across the lifespan contributes to mood fluctuations and cognitive dysfunction, two neurobehavioral features also associated with Alzheimer's Disease and Related Dementias (ADRD). Yet, few studies have used rodent models to determine how a history of ethanol consumption across the lifespan might contribute to neurobehavioral and neuropathological features of ADRD. We exposed Wild Type (WT) and transgenic Fischer 344 CE rats (TgF344-AD) that have been genetically modified to express the human Amyloid Precursor Protein (APP) and presenilin-1 genes with mutations, to ethanol using a chronic, intermittent ethanol consumption model. Beginning at P28, rats were given a single bottle 10 % ethanol solution for 2 consecutive days, followed by 2 days of tap water. This pattern (2 days on, days off) was repeated for a total of 12 cycles until rats reached the age of ∼ 3 months, and repeated at 6 (Exp 1 and Exp 2) and 9 months of age (Exp 2). In experiment 1, ethanol consumption decreased alternations in a spontaneous alternation task in females, only at the 3-month time point, whereas TgF344-AD females showed increased contextual fear conditioning in the test of retention and reinstatement tests at 6 months of age. In experiment 2, a battery of anxiety-like behaviors (Elevated Plus Maze, Marble Burying, and Novelty Induced Hypophagia) were assessed following a 2-week abstinence period at 3, 6, and 9 months of age in ethanol-consuming rats. Data from the EPM and marble burying tasks revealed evidence of heightened anxiety-like behavior in Tg-F344-AD rats that varied by sex and age, with no significant effects of ethanol. In the novelty-induced hypophagia task, males with a history of ethanol consumption had a lower latency to approach a familiar, salient reward at 3 months old, but effects of ethanol were overall minimal. Examination of dorsal hippocampal gene expression at 6 months of age under basal conditions also revealed predominantly genotype and sex-specific effects on inflammation- and AD-related genes (App, Il-6, Bace1, Rage, Lrp-1). When examined at 9 months old following LPS challenge, ethanol increased inflammatory genes in males (Il-1β, Il-6) in the hippocampus, whereas ethanol decreased several inflammatory and AD-related genes (Hmgb1, Rage, Bace1, Lrp-1) in TgF344-AD females. Overall, these data provide further evidence that females are especially vulnerable to AD, and that a history of ethanol consumption had selective, rather than global, effects on AD- and inflammation-related genes following an inflammatory stimulus.
Collapse
Affiliation(s)
- Paige Marsland
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Andrew S Vore
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Ashley Lutzke
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Anny Gano
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Abigail Fischer
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Sarah Trapp
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Lisa M Savage
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Terrence Deak
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States.
| |
Collapse
|
4
|
Ohene Y, Morrey WJ, Powell E, Smethers KF, Luka N, South K, Berks M, Lawrence CB, Parker GJM, Parkes LM, Boutin H, Dickie BR. MRI detects blood-brain barrier alterations in a rat model of Alzheimer's disease and lung infection. NPJ IMAGING 2025; 3:8. [PMID: 40051735 PMCID: PMC11879872 DOI: 10.1038/s44303-025-00071-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 01/23/2025] [Indexed: 03/09/2025]
Abstract
Pneumonia is a common infection in people suffering with Alzheimer's disease, leading to delirium, critical illness or severe neurological decline, which may be due to an amplified response of the blood-brain barrier (BBB) to peripheral insult. We assess the response of the BBB to repeated Streptococcus pneumoniae lung infection in rat model of Alzheimer's disease (TgF344-AD), at 13- and 18-months old, using dynamic contrast-enhanced (DCE) MRI and filter exchange imaging. Higher BBB water exchange rate is initially detected in infected TgF344-AD rats. BBB water exchange rates correlated with hippocampus aquaporin-4 water channel expression in infected animals. We detected no differences in BBB permeability to gadolinium contrast agent measured by DCE-MRI, confirmed by staining for tight junction proteins, occludin and claudin-5. These findings provide insight into the mechanisms of how peripheral inflammation impacts the BBB.
Collapse
Affiliation(s)
- Yolanda Ohene
- Division of Psychology, Communication and Human Neuroscience, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - William J. Morrey
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Elizabeth Powell
- Department of Medical Physics and Biomedical Engineering and Department of Neuroinflammation, Centre for Medical Image Computing, UCL, London, UK
| | - Katherine F. Smethers
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Nadim Luka
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Kieron South
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Michael Berks
- Division of Informatics, Imaging and Data Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Catherine B. Lawrence
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Geoff. J. M. Parker
- Department of Medical Physics and Biomedical Engineering and Department of Neuroinflammation, Centre for Medical Image Computing, UCL, London, UK
- Bioxydyn Limited, Manchester, UK
| | - Laura M. Parkes
- Division of Psychology, Communication and Human Neuroscience, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Hervé Boutin
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Imaging Brain & Neuropsychiatry iBraiN, Université de Tours, INSERM, Tours, France
| | - Ben R. Dickie
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Division of Informatics, Imaging and Data Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
5
|
Futácsi A, Rusznák K, Szarka G, Völgyi B, Wiborg O, Czéh B. Quantification and correlation of amyloid-β plaque load, glial activation, GABAergic interneuron numbers, and cognitive decline in the young TgF344-AD rat model of Alzheimer's disease. Front Aging Neurosci 2025; 17:1542229. [PMID: 40013092 PMCID: PMC11860898 DOI: 10.3389/fnagi.2025.1542229] [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: 12/09/2024] [Accepted: 01/28/2025] [Indexed: 02/28/2025] Open
Abstract
Background Animal models of Alzheimer's disease (AD) are essential tools for investigating disease pathophysiology and conducting preclinical drug testing. In this study, we examined neuronal and glial alterations in the hippocampus and medial prefrontal cortex (mPFC) of young TgF344-AD rats and correlated these changes with cognitive decline and amyloid-β plaque load. Methods We compared TgF344-AD and non-transgenic littermate rats aged 7-8 months of age. We systematically quantified β-amyloid plaques, astrocytes, microglia, four different subtypes of GABAergic interneurons (calretinin-, cholecystokinin-, parvalbumin-, and somatostatin-positive neurons), and newly generated neurons in the hippocampus. Spatial learning and memory were assessed using the Barnes maze test. Results Young TgF344-AD rats had a large number of amyloid plaques in both the hippocampus and mPFC, together with a pronounced increase in microglial cell numbers. Astrocytic activation was significant in the mPFC. Cholecystokinin-positive cell numbers were decreased in the hippocampus of transgenic rats, but calretinin-, parvalbumin-, and somatostatin-positive cell numbers were not altered. Adult neurogenesis was not affected by genotype. TgF344-AD rats had spatial learning and memory impairments, but this cognitive deficit did not correlate with amyloid plaque number or cellular changes in the brain. In the hippocampus, amyloid plaque numbers were negatively correlated with cholecystokinin-positive neuron and microglial cell numbers. In the mPFC, amyloid plaque number was negatively correlated with the number of astrocytes. Conclusion Pronounced neuropathological changes were found in the hippocampus and mPFC of young TgF344-AD rats, including the loss of hippocampal cholecystokinin-positive interneurons. Some of these neuropathological changes were negatively correlated with amyloid-β plaque load, but not with cognitive impairment.
Collapse
Affiliation(s)
- Anett Futácsi
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
- Imaging Core Facility, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Kitti Rusznák
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Gergely Szarka
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Imaging Core Facility, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Neurobiology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - Béla Völgyi
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Neurobiology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - Ove Wiborg
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Boldizsár Czéh
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
- Imaging Core Facility, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| |
Collapse
|
6
|
Morrey WJ, Ceyzériat K, Amossé Q, Badina AM, Dickie B, Schiessl I, Tsartsalis S, Millet P, Boutin H, Tournier BB. Early metabolic changes in the brain of Alzheimer's disease rats are driven by GLAST+ cells. J Cereb Blood Flow Metab 2025:271678X251318923. [PMID: 39917849 PMCID: PMC11806453 DOI: 10.1177/0271678x251318923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 11/27/2024] [Accepted: 01/19/2025] [Indexed: 02/11/2025]
Abstract
Glucose metabolic dysfunction is a hallmark of Alzheimer's disease (AD) pathology and is used to diagnose the disease or predict imminent cognitive decline. The main method to measure brain metabolism in vivo is positron emission tomography with 2-Deoxy-2-[18F]fluoroglucose ([18F]FDG-PET). The cellular origin of changes in the [18F]FDG-PET signal in AD is controversial. We addressed this by combining [18F]FDG-PET with subsequent cell-sorting and γ-counting of [18F]FDG-accumulation in sorted cell populations. 7-month-old male TgF344-AD rats and wild-type controls (n = 24/group) received sham or ceftriaxone (200 mg/kg) injection prior to [18F]FDG-PET imaging to increase glutamate uptake and glucose utilisation. The same animals were injected again one week later, and radiolabelled brains were dissected, with hippocampi taken for magnetically-activated cell sorting of radioligand-treated tissues (MACS-RTT). Radioactivity in sorted cell populations was measured to quantify cell-specific [18F]FDG uptake. Transcriptional analyses of metabolic enzymes/transporters were also performed. Hypometabolism in the frontal association cortex of TgF344-AD rats was identified using [18F]FDG-PET, whereas hypermetabolism was identified in the hippocampus using MACS-RTT. Hypermetabolism was primarily driven by GLAST+ cells. This was supported by transcriptional analyses which showed alteration to metabolic apparatus, including upregulation of hexokinase 2 and altered expression of glucose/lactate transporters. See Figure 1 for summary.
Collapse
Affiliation(s)
- William J Morrey
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Kelly Ceyzériat
- CIBM Center for BioMedical Imaging, Geneva, Switzerland
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
- Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Quentin Amossé
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
- Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | | | - Ben Dickie
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Ingo Schiessl
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Stergios Tsartsalis
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
- Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Philippe Millet
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
- Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Hervé Boutin
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
- Imaging Brain & Neuropsychiatry iBraiN U1253, Université de Tours, Inserm, Tours, France
| | - Benjamin B Tournier
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
- Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
7
|
Mohammad Hosseini A, Khaleghzadeh-Ahangar H, Rahimi A. The immunomodulatory effects of psychedelics in Alzheimer's disease-related dementia. Neuroscience 2025; 564:271-280. [PMID: 39603407 DOI: 10.1016/j.neuroscience.2024.11.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/03/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024]
Abstract
Dementia is an increasing disorder, and Alzheimer's disease (AD) is the cause of 60% of all dementia cases. Despite all efforts, there is no cure for stopping dementia progression. Recent studies reported potential effects of psychedelics on neuroinflammation during AD. Psychedelics by 5HT2AR activation can reduce proinflammatory cytokine levels (TNF-α, IL-6) and inhibit neuroinflammation. In addition to neuroinflammation suppression, psychedelics induce neuroplasticity by increasing Brain-derived neurotrophic factor (BDNF) levels through Sigma-1R stimulation. This review discussed the effects of psychedelics on AD from both neuroinflammatory and neuroplasticity standpoints.
Collapse
Affiliation(s)
| | - Hossein Khaleghzadeh-Ahangar
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Physiology, School of Medicine, Babol University of Medical Sciences, Babol, Iran; Mobility Impairment Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Atena Rahimi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Pharmacology and Toxicology, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
8
|
Wang H, Shi C, Jiang L, Liu X, Tang R, Tang M. Neuroimaging techniques, gene therapy, and gut microbiota: frontier advances and integrated applications in Alzheimer's Disease research. Front Aging Neurosci 2024; 16:1485657. [PMID: 39691161 PMCID: PMC11649678 DOI: 10.3389/fnagi.2024.1485657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disorder marked by cognitive decline, for which effective treatments remain elusive due to complex pathogenesis. Recent advances in neuroimaging, gene therapy, and gut microbiota research offer new insights and potential intervention strategies. Neuroimaging enables early detection and staging of AD through visualization of biomarkers, aiding diagnosis and tracking of disease progression. Gene therapy presents a promising approach for modifying AD-related genetic expressions, targeting amyloid and tau pathology, and potentially repairing neuronal damage. Furthermore, emerging evidence suggests that the gut microbiota influences AD pathology through the gut-brain axis, impacting inflammation, immune response, and amyloid metabolism. However, each of these technologies faces significant challenges, including concerns about safety, efficacy, and ethical considerations. This article reviews the applications, advantages, and limitations of neuroimaging, gene therapy, and gut microbiota research in AD, with a particular focus on their combined potential for early diagnosis, mechanistic insights, and therapeutic interventions. We propose an integrated approach that leverages these tools to provide a multi-dimensional framework for advancing AD diagnosis, treatment, and prevention.
Collapse
Affiliation(s)
- Haitao Wang
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Chen Shi
- Department of Gynaecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ling Jiang
- Department of Anorectal, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Xiaozhu Liu
- Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Rui Tang
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Mingxi Tang
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, Yaan People’s Hospital (Yaan Hospital of West China Hospital of Sichuan University), Yaan, Sichuan, China
| |
Collapse
|
9
|
Li Y, Liu Y, He Z, Li Z, Xiang H. Circadian Alterations in Brain Metabolism Linked to Cognitive Deficits During Hepatic Ischemia-Reperfusion Injury Using [ 1H- 13C]-NMR Metabolomics. Biomedicines 2024; 12:2536. [PMID: 39595102 PMCID: PMC11592224 DOI: 10.3390/biomedicines12112536] [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: 09/25/2024] [Revised: 10/26/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Hepatic ischemia-reperfusion injury (HIRI) is known to affect cognitive functions, with particular concern for its impact on brain metabolic dynamics. Circadian rhythms, as a crucial mechanism for internal time regulation within organisms, significantly influence metabolic processes in the brain. This study aims to explore how HIRI affects hippocampal metabolism and its circadian rhythm differences in mice, and to analyze how these changes are associated with cognitive impairments. Methods: A C57BL/6 male mouse model was used, simulating HIRI through hepatic ischemia-reperfusion surgery, with a sham operation conducted for the control group. Cognitive functions were evaluated using open field tests, Y-maze tests, and novel object recognition tests. Magnetic resonance spectroscopic imaging (MRSI) technology, combined with intravenous injection of [2-13C]-acetate and [1-13C]-glucose, was utilized to analyze metabolic changes in the hippocampus of HIRI mice at different circadian time points (Zeitgeber Time ZT0, 8:00 and ZT12, 20:00). Circadian rhythms regulate behavioral, physiological, and metabolic rhythms through transcriptional feedback loops, with ZT0 at dawn (lights on) and ZT12 at dusk (lights off). Results: HIRI mice exhibited significant cognitive impairments in behavioral tests, particularly in spatial memory and learning abilities. MRSI analysis revealed significant circadian rhythm differences in the concentration of metabolites in the hippocampus, with the enrichment concentrations of lactate, alanine, glutamate, and taurine showing different trends at ZT0 compared to ZT12, highlighting the important influence of circadian rhythms on metabolic dysregulation induced by HIRI. Conclusions: This study highlights the significant impact of HIRI on brain metabolic dynamics in mice, especially in the hippocampal area, and for the first time reveals the differences in these effects within circadian rhythms. These findings not only emphasize the association between HIRI-induced cognitive impairments and changes in brain metabolism but also point out the crucial role of circadian rhythms in this process, offering new metabolic targets and timing considerations for therapeutic strategies against HIRI-related cognitive disorders.
Collapse
Affiliation(s)
- Yijing Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.L.); (Y.L.); (Z.H.)
| | - Yanbo Liu
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.L.); (Y.L.); (Z.H.)
| | - Zhigang He
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.L.); (Y.L.); (Z.H.)
| | - Zhixiao Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.L.); (Y.L.); (Z.H.)
| | - Hongbing Xiang
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.L.); (Y.L.); (Z.H.)
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430030, China
| |
Collapse
|
10
|
Almanza DLV, Koletar MM, Lai AY, Lam WW, Joo L, Hill ME, Stanisz GJ, McLaurin J, Stefanovic B. High caloric intake improves neuronal metabolism and functional hyperemia in a rat model of early AD pathology. Theranostics 2024; 14:7405-7423. [PMID: 39659583 PMCID: PMC11626934 DOI: 10.7150/thno.98793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 10/24/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction: While obesity has been linked to both increased and decreased rate of cognitive decline in Alzheimer's Disease (AD) patients, there is no consensus on the interaction between obesity and AD. Methods: The TgF344-AD rat model was used to investigate the effects of high carbohydrate, high fat (HCHF) diet on brain glucose metabolism and hemodynamics in the presence or absence of AD transgenes, in presymptomatic (6-month-old) vs. symptomatic (12-month-old) stages of AD progression using non-invasive neuroimaging. Results: In presymptomatic AD, HCHF exerted detrimental effects, attenuating both hippocampal glucose uptake and resting perfusion in both non-transgenic and TgAD cohorts, when compared to CHOW-fed cohorts. In contrast, HCHF consumption was beneficial in established AD, resolving the AD-progression associated attenuation in hippocampal glucose uptake and functional hyperemia. Discussion: Whereas HCHF was harmful to the presymptomatic AD brain, it ameliorated deficits in hippocampal metabolism and neurovascular coupling in symptomatic TgAD rats.
Collapse
Affiliation(s)
- Dustin Loren V. Almanza
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | | | - Aaron Y. Lai
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Wilfred W. Lam
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Lewis Joo
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Mary E. Hill
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Greg J. Stanisz
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University, Lublin, Poland
| | - JoAnne McLaurin
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Bojana Stefanovic
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|
11
|
Kong Y, Maschio CA, Shi X, Xie F, Zuo C, Konietzko U, Shi K, Rominger A, Xiao J, Huang Q, Nitsch RM, Guan Y, Ni R. Relationship Between Reactive Astrocytes, by [ 18F]SMBT-1 Imaging, with Amyloid-Beta, Tau, Glucose Metabolism, and TSPO in Mouse Models of Alzheimer's Disease. Mol Neurobiol 2024; 61:8387-8401. [PMID: 38502413 DOI: 10.1007/s12035-024-04106-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 03/06/2024] [Indexed: 03/21/2024]
Abstract
Reactive astrocytes play an important role in the development of Alzheimer's disease (AD). Here, we aimed to investigate the temporospatial relationships among monoamine oxidase-B, tau and amyloid-β (Aβ), translocator protein, and glucose metabolism by using multitracer imaging in AD transgenic mouse models. Positron emission tomography (PET) imaging with [18F]SMBT-1 (monoamine oxidase-B), [18F]florbetapir (Aβ), [18F]PM-PBB3 (tau), [18F]fluorodeoxyglucose (FDG), and [18F]DPA-714 (translocator protein) was carried out in 5- and 10-month-old APP/PS1, 11-month-old 3×Tg mice, and aged-matched wild-type mice. The brain regional referenced standard uptake value (SUVR) was computed with the cerebellum as the reference region. Immunofluorescence staining was performed on mouse brain tissue slices. [18F]SMBT-1 and [18F]florbetapir SUVRs were greater in the cortex and hippocampus of 10-month-old APP/PS1 mice than in those of 5-month-old APP/PS1 mice and wild-type mice. No significant difference in the regional [18F]FDG or [18F]DPA-714 SUVRs was observed in the brains of 5- or 10-month-old APP/PS1 mice or wild-type mice. No significant difference in the SUVRs of any tracer was observed between 11-month-old 3×Tg mice and age-matched wild-type mice. A positive correlation between the SUVRs of [18F]florbetapir and [18F]DPA-714 in the cortex and hippocampus was observed among the transgenic mice. Immunostaining validated the distribution of MAO-B and limited Aβ and tau pathology in 11-month-old 3×Tg mice; and Aβ deposits in brain tissue from 10-month-old APP/PS1 mice. In summary, these findings provide in vivo evidence that an increase in astrocyte [18F]SMBT-1 accompanies Aβ accumulation in APP/PS1 models of AD amyloidosis.
Collapse
Affiliation(s)
- Yanyan Kong
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Cinzia A Maschio
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Zurich Neuroscience Zentrum (ZNZ), Zurich, Switzerland
| | - Xuefeng Shi
- Qinghai Provincial People's Hospital, Xining, China
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Chuantao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Uwe Konietzko
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, University of Bern, Bern, Switzerland
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, University of Bern, Bern, Switzerland
| | - Jianfei Xiao
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qi Huang
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Roger M Nitsch
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China.
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.
- Zurich Neuroscience Zentrum (ZNZ), Zurich, Switzerland.
- Department of Nuclear Medicine, Inselspital, University of Bern, Bern, Switzerland.
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
12
|
Soyer A, Goutal S, Leterrier S, Marie S, Larrat B, Selingue E, Winkeler A, Sarazin M, Bottlaender M, Tournier N. [ 18F]2-fluoro-2-deoxy-sorbitol ([ 18F]FDS) PET imaging repurposed for quantitative estimation of blood-brain barrier permeability in a rat model of Alzheimer's disease. ANNALES PHARMACEUTIQUES FRANÇAISES 2024; 82:822-829. [PMID: 38657857 DOI: 10.1016/j.pharma.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/05/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024]
Abstract
Numerous studies suggest that blood-brain barrier (BBB) dysfunction may contribute to the progression of Alzheimer's disease (AD). Clinically available neuroimaging methods are needed for quantitative "scoring" of BBB permeability in AD patients. [18F]2-fluoro-2-deoxy-sorbitol ([18F]FDS), which can be easily obtained from simple chemical reduction of commercial [18F]2-fluoro-2-deoxy-glucose ([18F]FDG), was investigated as a small-molecule marker of BBB permeability, in a pre-clinical model of AD using in vivo PET imaging. Chemical reduction of [18F]FDG to [18F]FDS was obtained with a 100% conversion yield. Dynamic PET acquisitions were performed in the APP/PS1 rat model of AD (TgF344-AD, n=3) compared with age-matched littermates (WT, n=4). The brain uptake of [18F]FDS was determined in selected brain regions, delineated from a coregistered rat brain template. The brain uptake of [18F]FDS in the brain regions of AD rats versus WT rats was compared using a 2-way ANOVA. The uptake of [18F]FDS was significantly higher in the whole brain of AD rats, as compared with WT rats (P<0.001), suggesting increased BBB permeability. Enhanced brain uptake of [18F]FDS in AD rats was significantly different across brain regions (P<0.001). Minimum difference was observed in the amygdala (+89.0±7.6%, P<0.001) and maximum difference was observed in the midbrain (+177.8±29.2%, P<0.001). [18F]FDS, initially proposed as radio-pharmaceutical to estimate renal filtration using PET imaging, can be repurposed for non-invasive and quantitative determination of BBB permeability in vivo. Making the best with the quantitative properties of PET imaging, it was possible to estimate the extent of enhanced BBB permeability in a rat model of AD.
Collapse
Affiliation(s)
- Amélie Soyer
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Sébastien Goutal
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Sarah Leterrier
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Solène Marie
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Benoit Larrat
- Centre d'études de Saclay, CEA, CNRS, NeuroSpin/BAOBAB, Paris-Saclay University, 91191 Gif-sur-Yvette, France
| | - Erwan Selingue
- Centre d'études de Saclay, CEA, CNRS, NeuroSpin/BAOBAB, Paris-Saclay University, 91191 Gif-sur-Yvette, France
| | - Alexandra Winkeler
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Marie Sarazin
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Michel Bottlaender
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Nicolas Tournier
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France.
| |
Collapse
|
13
|
Reitz NL, Nunes PT, Savage LM. Adolescent alcohol exposure alters age-related progression of behavioral and neurotrophic dysfunction in the TgF344-AD model in a sex-specific manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.17.603911. [PMID: 39091885 PMCID: PMC11291002 DOI: 10.1101/2024.07.17.603911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Alzheimer's Disease (AD) and heavy alcohol use are widely prevalent and lead to brain pathology. Both alcohol-related brain damage (ABRD) and AD result in cholinergic dysfunction, reductions in hippocampal neurogenesis, and the emergence of hippocampal-dependent cognitive impairments. It is still unknown how ARBD caused during a critical developmental timepoint, such as adolescence, interacts with AD-related pathologies to accelerate disease progression later in life. The current study utilized a longitudinal design to characterize behavioral and pathological changes in a transgenic rat model of AD (TgF344-AD) following adolescent intermittent ethanol (AIE) exposure. We found that AIE accelerates cognitive decline associated with AD transgenes in female rats at 6 months of age, and male AD-rats are impaired on spatial navigation by 3-months with no additional deficits due to AIE exposure. Protein levels of various AD-pathological markers were analyzed in the dorsal and ventral hippocampus of male and female rats. The data suggests that AIE-induced alterations of the tropomyosin-related kinase A receptor (TrkA) / p75 neurotrophin receptor (p75NTR) ratio creates a brain that is vulnerable to age- and AD-related pathologies, which leads to an acceleration of cognitive decline, particularly in female rats.
Collapse
|
14
|
Kong Y, Cao L, Wang J, Zhuang J, Xie F, Zuo C, Huang Q, Shi K, Rominger A, Li M, Wu P, Guan Y, Ni R. In vivo reactive astrocyte imaging using [ 18F]SMBT-1 in tauopathy and familial Alzheimer's disease mouse models: A multi-tracer study. J Neurol Sci 2024; 462:123079. [PMID: 38878650 DOI: 10.1016/j.jns.2024.123079] [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: 01/24/2024] [Revised: 05/13/2024] [Accepted: 06/03/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Reactive astrocytes play an important role in the development of Alzheimer's disease and primary tauopathies. Here, we aimed to investigate the relationships between reactive astrocytes. Microgliosis and glucose metabolism with Tau and amyloid beta pathology by using multi-tracer imaging in widely used tauopathy and familial Alzheimer's disease mouse models. RESULTS Positron emission tomography imaging using [18F]PM-PBB3 (tau), [18F]florbetapir (amyloid-beta), [18F]SMBT-1 (monoamine oxidase-B), [18F]DPA-714 (translocator protein) and [18F]fluorodeoxyglucose was carried out in 3- and 7-month-old rTg4510 tau mice, 5 × FAD familial Alzheimer's disease mice and wild-type mice. Immunofluorescence staining was performed to validate the pathological distribution in the mouse brain after in vivo imaging. We found increased regional levels of [18F]PM-PBB3, [18F]SMBT-1, and [18F]DPA-714 and hypoglucose metabolism in the brains of 7-month-old rTg4510 mice compared to age-matched wild-type mice. Increased [18F]SMBT-1 uptake was observed in the brains of 3, 7-month-old 5 × FAD mice, with elevated regional [18F]florbetapir and [18F]DPA-714 uptakes in the brains of 7-month-old 5 × FAD mice, compared to age-matched wild-type mice. Positive correlations were shown between [18F]SMBT-1 and [18F]PM-PBB3, [18F]DPA-714 and [18F]PM-PBB3 in rTg4510 mice, and between [18F]florbetapir and [18F]DPA-714 SUVRs in 5 × FAD mice. CONCLUSION In summary, these findings provide in vivo evidence that reactive astrocytes, microglial activation, and cerebral hypoglucose metabolism are associated with tau and amyloid pathology development in animal models of tauopathy and familial Alzheimer's disease.
Collapse
Affiliation(s)
- Yanyan Kong
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Lei Cao
- PET Center, Huashan Hospital, Fudan University, Shanghai, China; Inst. Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Jiao Wang
- Lab of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Junyi Zhuang
- Lab of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Chuantao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qi Huang
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Kuangyu Shi
- Dept. Nuclear Medicine, Bern University Hospital, Bern, Switzerland
| | - Axel Rominger
- Dept. Nuclear Medicine, Bern University Hospital, Bern, Switzerland
| | - Ming Li
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Ping Wu
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China.
| | - Ruiqing Ni
- Inst. Regenerative Medicine, University of Zurich, Zurich, Switzerland; Dept. Nuclear Medicine, Bern University Hospital, Bern, Switzerland; Inst. Biomedical Engineering, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
15
|
Zeng H, Zhang Q, Liu L, Deng F, Han H, Meng F, Bai H. Correlation between abnormal cellular immune and changes of magnetic resonance spectroscopy in patients with Alzheimer's disease. Neurochem Int 2024; 176:105737. [PMID: 38599243 DOI: 10.1016/j.neuint.2024.105737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/27/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024]
Abstract
BACKGROUND Evidence from previous studies indicates that neuroinflammation contributes to the onset of Alzheimer's Disease (AD). Moreover, cellular dysfunction is induced by impaired signaling of neurotransmitters. This study aimed to explore the correlation between cellular immune dysfunction and neurotransmitter changes through cranial Magnetic Resonance Spectroscopy (MRS) in AD patients. METHODS Here, 32 AD, 40 Vascular Dementia (VD), and 35 Non-Dementia Elderly Control (NDE) cases were enrolled. Flow cytometry was performed to characterize lymphocyte subsets in plasma samples. The IL-1β and Caspase-1 levels were detected by ELISA. The NLRP3 expression level was measured by Western Blot (WB). The equivalence of N-acetylaspartate (NAA), Creatine (Cr), Choline (Cho), and Inositol (MI) in bilateral hippocampi of patients was examined by MRS. The association of NAA/Cr or MI/Cr ratios with the proportion of T lymphocyte subsets or NK cell subsets was determined through single-factor correlation analysis. RESULTS The proportion of T lymphocyte subsets was significantly lower in the AD group than in the NDE group (P < 0.01). On the other hand, the Caspase-1, NLRP3, and IL-1β protein expression levels were significantly higher in the AD group than in the other groups. Further analysis showed that the NAA/Cr ratio was lower in the AD group than in the NDE group. Additionally, a significant positive correlation was found between the NAA/Cr ratio and the MMSE score (r = 0.81, P < 0.01). Moreover, a significant positive correlation was observed between the NAA/Cr and T lymphocyte ratios. The NAA/Cr ratio was significantly negatively correlated with the proportion of NK cells in the blood (r = -0.83, P < 0.01). A significant negative correlation was also recorded between the MI/Cr and T cell ratios in blood samples. CONCLUSIONS Impaired cellular immune dysfunction in AD patients was significantly correlated with abnormal MRS. Neuroimmune dysfunction may contribute to the pathogenesis of AD and alter the metabolism of neurotransmitters such as aspartic acid and MI in the brains of AD patients. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
- Hongmei Zeng
- Department of Neurology, The Third Affiliated Hospital of Guizhou Medical University, Duyun, 558099, China; Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Qifang Zhang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, 550004, China; Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, 550004, China
| | - Lijie Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050004, China
| | - Feifei Deng
- Department of Neurology, The Third Affiliated Hospital of Guizhou Medical University, Duyun, 558099, China
| | - Huabo Han
- Department of Radiology, The Third Affiliated Hospital of Guizhou Medical University, Duyun, 558099, China
| | - Fuxue Meng
- Medical Laboratory Center, Third Affiliated Hospital of Guizhou Medical University, Duyun, 558099, China
| | - Hua Bai
- Department of Neurology, The Third Affiliated Hospital of Guizhou Medical University, Duyun, 558099, China; Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China; Medical Laboratory Center, Third Affiliated Hospital of Guizhou Medical University, Duyun, 558099, China.
| |
Collapse
|
16
|
Xu M, Liu J, Liu Q, Gong Y, Li Y, Zhang J, Shi S, Shi Y. Preliminary study on early diagnosis of Alzheimer's disease in APP/PS1 transgenic mice using multimodal magnetic resonance imaging. Front Aging Neurosci 2024; 16:1326394. [PMID: 38419647 PMCID: PMC10899441 DOI: 10.3389/fnagi.2024.1326394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/22/2024] [Indexed: 03/02/2024] Open
Abstract
Alzheimer's disease (AD) has an insidious onset and lacks clear early diagnostic markers, and by the time overt dementia symptoms appear, the disease is already in the mid-to-late stages. The search for early diagnostic markers of AD may open a critical window for Alzheimer's treatment and facilitate early intervention to slow the progression of AD. In this study, we aimed to explore the imaging markers for early diagnosis of AD through the combined application of structural magnetic resonance imaging (sMRI), resting-state functional magnetic resonance imaging (rs-fMRI), and 1H-magnetic resonance spectroscopy (1H-MRS) multimodal magnetic resonance imaging (MRI) techniques at the animal experimental level, with the aim to provide a certain reference for early clinical diagnosis of AD. First, sMRI scans were performed on 4-month-old amyloid beta precursor protein/presenilin 1 (APP/PS1) transgenic AD model mice and wild type mice of the same litter using a 7.0 T animal MRI scanner to analyze the differential brain regions with structural changes in the gray matter of the brain by voxel-based morphometry (VBM). Next, rs-fMRI scans were performed to analyze the differential brain regions between groups for local spontaneous brain activity and functional connectivity (FC) between brain regions. Finally, 1H-MRS scans were performed to quantify and analyze intergroup differences in the relative concentrations of different metabolites within regions of interest (cortex and hippocampus). Compared with wild type mice, the volume of the left hippocampus, and right olfactory bulb of APP/PS1 transgenic AD model mice were reduced, the functional activity of the bilateral hippocampus, right piriform cortex and right caudate putamen was reduced, the functional network connectivity of the hippocampus was impaired, and the relative content of N-acetylaspartate (NAA)in the hippocampus was decreased. In addition, this study found that imaging changes in olfactory-related brain regions were closely associated with AD diagnosis, and these findings may provide some reference for the early diagnosis of AD.
Collapse
Affiliation(s)
- Meng Xu
- Department of Tuina, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Jipeng Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Qingguo Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Gong
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yinyin Li
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
- Department Shenzhen Hospital (Longgang), Beijing University of Chinese Medicine, Shenzhen, China
| | - Jing Zhang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Shufeng Shi
- Department of Tuina, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Yuanyuan Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
- Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China
- Shenzhen Cell Valley Biopharmaceuticals Co., Ltd., Shenzhen, China
| |
Collapse
|
17
|
Liu Y, Tan Y, Zhang Z, Yi M, Zhu L, Peng W. The interaction between ageing and Alzheimer's disease: insights from the hallmarks of ageing. Transl Neurodegener 2024; 13:7. [PMID: 38254235 PMCID: PMC10804662 DOI: 10.1186/s40035-024-00397-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/31/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Ageing is a crucial risk factor for Alzheimer's disease (AD) and is characterised by systemic changes in both intracellular and extracellular microenvironments that affect the entire body instead of a single organ. Understanding the specific mechanisms underlying the role of ageing in disease development can facilitate the treatment of ageing-related diseases, such as AD. Signs of brain ageing have been observed in both AD patients and animal models. Alleviating the pathological changes caused by brain ageing can dramatically ameliorate the amyloid beta- and tau-induced neuropathological and memory impairments, indicating that ageing plays a crucial role in the pathophysiological process of AD. In this review, we summarize the impact of several age-related factors on AD and propose that preventing pathological changes caused by brain ageing is a promising strategy for improving cognitive health.
Collapse
Affiliation(s)
- Yuqing Liu
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China
| | - Yejun Tan
- School of Mathematics, University of Minnesota Twin Cities, Minneapolis, MN, 55455, USA
| | - Zheyu Zhang
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China
| | - Min Yi
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China
| | - Lemei Zhu
- Academician Workstation, Changsha Medical University, Changsha, 410219, People's Republic of China
| | - Weijun Peng
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China.
- National Clinical Research Center for Metabolic Diseases, Changsha, 410011, People's Republic of China.
| |
Collapse
|
18
|
Rudisch DM, Krasko MN, Barnett DGS, Mueller KD, Russell JA, Connor NP, Ciucci MR. Early ultrasonic vocalization deficits and related thyroarytenoid muscle pathology in the transgenic TgF344-AD rat model of Alzheimer's disease. Front Behav Neurosci 2024; 17:1294648. [PMID: 38322496 PMCID: PMC10844490 DOI: 10.3389/fnbeh.2023.1294648] [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: 09/15/2023] [Accepted: 12/01/2023] [Indexed: 02/08/2024] Open
Abstract
Background Alzheimer's disease (AD) is a progressive neurologic disease and the most common cause of dementia. Classic pathology in AD is characterized by inflammation, abnormal presence of tau protein, and aggregation of β-amyloid that disrupt normal neuronal function and lead to cell death. Deficits in communication also occur during disease progression and significantly reduce health, well-being, and quality of life. Because clinical diagnosis occurs in the mid-stage of the disease, characterizing the prodrome and early stages in humans is currently challenging. To overcome these challenges, we use the validated TgF344-AD (F344-Tg(Prp-APP, Prp-PS1)19/Rrrc) transgenic rat model that manifests cognitive, behavioral, and neuropathological dysfunction akin to AD in humans. Objectives The overarching goal of our work is to test the central hypothesis that pathology and related behavioral deficits such as communication dysfunction in part manifest in the peripheral nervous system and corresponding target tissues already in the early stages. The primary aims of this study are to test the hypotheses that: (1) changes in ultrasonic vocalizations (USV) occur in the prodromal stage at 6 months of age and worsen at 9 months of age, (2) inflammation as well as AD-related pathology can be found in the thyroarytenoid muscle (TA) at 12 months of age (experimental endpoint tissue harvest), and to (3) demonstrate that the TgF344-AD rat model is an appropriate model for preclinical investigations of early AD-related vocal deficits. Methods USVs were collected from male TgF344-AD (N = 19) and wildtype (WT) Fischer-344 rats (N = 19) at 6 months (N = 38; WT: n = 19; TgF344-AD: n = 19) and 9 months of age (N = 18; WT: n = 10; TgF344-AD: n = 8) and acoustically analyzed for duration, mean power, principal frequency, low frequency, high frequency, peak frequency, and call type. RT-qPCR was used to assay peripheral inflammation and AD-related pathology via gene expressions in the TA muscle of male TgF344-AD rats (n = 6) and WT rats (n = 6) at 12 months of age. Results This study revealed a significant reduction in mean power of ultrasonic calls from 6 to 9 months of age and increased peak frequency levels over time in TgF344-AD rats compared to WT controls. Additionally, significant downregulation of AD-related genes Uqcrc2, Bace2, Serpina3n, and Igf2, as well as downregulation of pro-inflammatory gene Myd88 was found in the TA muscle of TgF344-AD rats at 12 months of age. Discussion Our findings demonstrate early and progressive vocal deficits in the TgF344-AD rat model. We further provide evidence of dysregulation of AD-pathology-related genes as well as inflammatory genes in the TA muscles of TgF344-AD rats in the early stage of the disease, confirming this rat model for early-stage investigations of voice deficits and related pathology.
Collapse
Affiliation(s)
- Denis Michael Rudisch
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, United States
- Department of Surgery, Division of Otolaryngology - Head and Neck Surgery, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- UW Institute for Clinical and Translational Research, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Maryann N Krasko
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, United States
- Department of Surgery, Division of Otolaryngology - Head and Neck Surgery, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - David G S Barnett
- Department of Surgery, Division of Otolaryngology - Head and Neck Surgery, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Kimberly D Mueller
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, United States
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - John A Russell
- Department of Surgery, Division of Otolaryngology - Head and Neck Surgery, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Nadine P Connor
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, United States
- Department of Surgery, Division of Otolaryngology - Head and Neck Surgery, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Michelle R Ciucci
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, United States
- Department of Surgery, Division of Otolaryngology - Head and Neck Surgery, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
19
|
Kumar S, Mahajan A, Ambatwar R, Khatik GL. Recent Advancements in the Treatment of Alzheimer's Disease: A Multitarget-directed Ligand Approach. Curr Med Chem 2024; 31:6032-6062. [PMID: 37861025 DOI: 10.2174/0109298673264076230921065945] [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: 05/25/2023] [Revised: 08/18/2023] [Accepted: 08/24/2023] [Indexed: 10/21/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease and one of the leading causes of progressive dementia, affecting 50 million people worldwide. Many pathogenic processes, including amyloid β aggregation, tau hyperphosphorylation, oxidative stress, neuronal death, and deterioration of the function of cholinergic neurons, are associated with its progression. The one-compound-one-target treatment paradigm was unsuccessful in treating AD due to the multifaceted nature of Alzheimer's disease. The recent development of multitarget-directed ligand research has been explored to target the complementary pathways associated with the disease. We aimed to find the key role and progress of MTDLs in treating AD; thus, we searched for the past ten years of literature on "Pub- Med", "ScienceDirect", "ACS" and "Bentham Science" using the keywords neurodegenerative diseases, Alzheimer's disease, and multitarget-directed ligands. The literature was further filtered based on the quality of work and relevance to AD. Thus, this review highlights the current advancement and advantages of multitarget-directed ligands over traditional single-targeted drugs and recent progress in their development to treat AD.
Collapse
Affiliation(s)
- Sumit Kumar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| | - Amol Mahajan
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| | - Ramesh Ambatwar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| | - Gopal L Khatik
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| |
Collapse
|
20
|
Sagalajev B, Lennartz L, Vieth L, Gunawan CT, Neumaier B, Drzezga A, Visser-Vandewalle V, Endepols H, Sesia T. TgF344-AD Rat Model of Alzheimer's Disease: Spatial Disorientation and Asymmetry in Hemispheric Neurodegeneration. J Alzheimers Dis Rep 2023; 7:1085-1094. [PMID: 37849636 PMCID: PMC10578321 DOI: 10.3233/adr-230038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/22/2023] [Indexed: 10/19/2023] Open
Abstract
Background The TgF344-AD ratline represents a transgenic animal model of Alzheimer's disease. We previously reported spatial memory impairment in TgF344-AD rats, yet the underlying mechanism remained unknown. We, therefore, set out to determine if spatial memory impairment in TgF344-AD rats is attributed to spatial disorientation. Also, we aimed to investigate whether TgF344-AD rats exhibit signs of asymmetry in hemispheric neurodegeneration, similar to what is reported in spatially disoriented AD patients. Finally, we sought to examine how spatial disorientation correlates with working memory performance. Methods TgF344-AD rats were divided into two groups balanced by sex and genotype. The first group underwent the delayed match-to-sample (DMS) task for the assessment of spatial orientation and working memory, while the second group underwent positron emission tomography (PET) for the assessment of glucose metabolism and microglial activity as in-vivo markers of neurodegeneration. Rats were 13 months old during DMS training and 14-16 months old during DMS testing and PET. Results In the DMS task, TgF344-AD rats were more likely than their wild-type littermates to display strong preference for one of the two levers, preventing working memory testing. Rats without lever-preference showed similar working memory, regardless of their genotype. PET revealed hemispherically asymmetric clusters of increased microglial activity and altered glucose metabolism in TgF344-AD rats. Conclusions TgF344-AD rats display spatial disorientation and hemispherically asymmetrical neurodegeneration, suggesting a potential causal relationship consistent with past clinical research. In rats with preserved spatial orientation, working memory remains intact.
Collapse
Affiliation(s)
- Boriss Sagalajev
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Stereotactic and Functional Neurosurgery, Cologne, Germany
- European Graduate School of Neuroscience (EURON), Maastricht, Netherlands
| | - Lina Lennartz
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Stereotactic and Functional Neurosurgery, Cologne, Germany
- European Graduate School of Neuroscience (EURON), Maastricht, Netherlands
| | - Lukas Vieth
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, Cologne, Germany
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Jülich, Germany
| | - Cecilia Tasya Gunawan
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Stereotactic and Functional Neurosurgery, Cologne, Germany
- European Graduate School of Neuroscience (EURON), Maastricht, Netherlands
| | - Bernd Neumaier
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, Cologne, Germany
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Jülich, Germany
| | - Alexander Drzezga
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Nuclear Medicine, Cologne, Germany
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Molecular Organization of the Brain (INM-2), Jülich, Germany
| | - Veerle Visser-Vandewalle
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Stereotactic and Functional Neurosurgery, Cologne, Germany
- European Graduate School of Neuroscience (EURON), Maastricht, Netherlands
| | - Heike Endepols
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, Cologne, Germany
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Jülich, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Nuclear Medicine, Cologne, Germany
| | - Thibaut Sesia
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Stereotactic and Functional Neurosurgery, Cologne, Germany
- European Graduate School of Neuroscience (EURON), Maastricht, Netherlands
| |
Collapse
|
21
|
Fang X, Tang C, Zhang H, Border JJ, Liu Y, Shin SM, Yu H, Roman RJ, Fan F. Longitudinal characterization of cerebral hemodynamics in the TgF344-AD rat model of Alzheimer's disease. GeroScience 2023; 45:1471-1490. [PMID: 36933144 PMCID: PMC10400494 DOI: 10.1007/s11357-023-00773-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 03/12/2023] [Indexed: 03/19/2023] Open
Abstract
Alzheimer's disease (AD) is a global healthcare crisis. The TgF344-AD rat is an AD model exhibiting age-dependent AD pathological hallmarks. We confirmed that AD rats developed cognitive deficits at 6 months without alteration of any other major biophysical parameters. We longitudinally characterized cerebral hemodynamics in AD rats at 3, 4, 6, and 14 months. The myogenic responses of the cerebral arteries and arterioles were impaired at 4 months of age in the AD rats. Consistent with the ex vivo results, the AD rat exhibited poor autoregulation of surface and deep cortical cerebral blood flow 2 months preceding cognitive decline. The dysfunction of cerebral hemodynamics in AD is exacerbated with age associated with reduced cerebral perfusion. Further, abolished cell contractility contributes to cerebral hemodynamics imbalance in AD. This may be attributed to enhanced ROS production, reduced mitochondrial respiration and ATP production, and disrupted actin cytoskeleton in cerebral vascular contractile cells.
Collapse
Affiliation(s)
- Xing Fang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Chengyun Tang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
- Department of Physiology, Medical College of Georgia, Augusta University, 1462 Laney Walker Blvd, Augusta, GA, 30912, USA
| | - Huawei Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Jane J Border
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Yedan Liu
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
- Department of Physiology, Medical College of Georgia, Augusta University, 1462 Laney Walker Blvd, Augusta, GA, 30912, USA.
| |
Collapse
|
22
|
Fontana IC, Kumar A, Nordberg A. The role of astrocytic α7 nicotinic acetylcholine receptors in Alzheimer disease. Nat Rev Neurol 2023; 19:278-288. [PMID: 36977843 DOI: 10.1038/s41582-023-00792-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 03/30/2023]
Abstract
The ongoing search for therapeutic interventions in Alzheimer disease (AD) has highlighted the complexity of this condition and the need for additional biomarkers, beyond amyloid-β (Aβ) and tau, to improve clinical assessment. Astrocytes are brain cells that control metabolic and redox homeostasis, among other functions, and are emerging as an important focus of AD research owing to their swift response to brain pathology in the initial stages of the disease. Reactive astrogliosis - the morphological, molecular and functional transformation of astrocytes during disease - has been implicated in AD progression, and the definition of new astrocytic biomarkers could help to deepen our understanding of reactive astrogliosis along the AD continuum. As we highlight in this Review, one promising biomarker candidate is the astrocytic α7 nicotinic acetylcholine receptor (α7nAChR), upregulation of which correlates with Aβ pathology in the brain of individuals with AD. We revisit the past two decades of research into astrocytic α7nAChRs to shed light on their roles in the context of AD pathology and biomarkers. We discuss the involvement of astrocytic α7nAChRs in the instigation and potentiation of early Aβ pathology and explore their potential as a target for future reactive astrocyte-based therapeutics and imaging biomarkers in AD.
Collapse
Affiliation(s)
- Igor C Fontana
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Amit Kumar
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
23
|
Yu R, Maswikiti EP, Yu Y, Gao L, Ma C, Ma H, Deng X, Wang N, Wang B, Chen H. Advances in the Application of Preclinical Models in Photodynamic Therapy for Tumor: A Narrative Review. Pharmaceutics 2023; 15:pharmaceutics15010197. [PMID: 36678826 PMCID: PMC9867105 DOI: 10.3390/pharmaceutics15010197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/29/2022] [Accepted: 01/01/2023] [Indexed: 01/09/2023] Open
Abstract
Photodynamic therapy (PDT) is a non-invasive laser light local treatment that has been utilized in the management of a wide variety of solid tumors. Moreover, the evaluation of efficacy, adverse reactions, the development of new photosensitizers and the latest therapeutic regimens are inseparable from the preliminary exploration in preclinical studies. Therefore, our aim was to better comprehend the characteristics and limitations of these models and to provide a reference for related research. METHODS We searched the databases, including PubMed, Web of Science and Scopus for the past 25 years of original research articles on the feasibility of PDT in tumor treatment based on preclinical experiments and animal models. We provided insights into inclusion and exclusion criteria and ultimately selected 40 articles for data synthesis. RESULTS After summarizing and comparing the methods and results of these studies, the experimental model selection map was drawn. There are 7 main preclinical models, which are used for different research objectives according to their characteristics. CONCLUSIONS Based on this narrative review, preclinical experimental models are crucial to the development and promotion of PDT for tumors. The traditional animal models have some limitations, and the emergence of organoids may be a promising new insight.
Collapse
Affiliation(s)
- Rong Yu
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | | | - Yang Yu
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Lei Gao
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Chenhui Ma
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Huanhuan Ma
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Xiaobo Deng
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Na Wang
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Bofang Wang
- The Second Clinical College of Medicine, Lanzhou University, Lanzhou 730030, China
| | - Hao Chen
- Department of Surgical Oncology, Second Hospital of Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Digestive System Tumor of Gansu Province, Second Hospital of Lanzhou University, Lanzhou 730030, China
- Correspondence: ; Tel.: +86-0931-5190550
| |
Collapse
|
24
|
Černotová D, Hrůzová K, Levčík D, Svoboda J, Stuchlík A. Linking Social Cognition, Parvalbumin Interneurons, and Oxytocin in Alzheimer's Disease: An Update. J Alzheimers Dis 2023; 96:861-875. [PMID: 37980658 PMCID: PMC10741376 DOI: 10.3233/jad-230333] [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] [Accepted: 09/11/2023] [Indexed: 11/21/2023]
Abstract
Finding a cure for Alzheimer's disease (AD) has been notoriously challenging for many decades. Therefore, the current focus is mainly on prevention, timely intervention, and slowing the progression in the earliest stages. A better understanding of underlying mechanisms at the beginning of the disease could aid in early diagnosis and intervention, including alleviating symptoms or slowing down the disease progression. Changes in social cognition and progressive parvalbumin (PV) interneuron dysfunction are among the earliest observable effects of AD. Various AD rodent models mimic these early alterations, but only a narrow field of study has considered their mutual relationship. In this review, we discuss current knowledge about PV interneuron dysfunction in AD and emphasize their importance in social cognition and memory. Next, we propose oxytocin (OT) as a potent modulator of PV interneurons and as a promising treatment for managing some of the early symptoms. We further discuss the supporting evidence on its beneficial effects on AD-related pathology. Clinical trials have employed the use of OT in various neuropsychiatric diseases with promising results, but little is known about its prospective impacts on AD. On the other hand, the modulatory effects of OT in specific structures and local circuits need to be clarified in future studies. This review highlights the connection between PV interneurons and social cognition impairment in the early stages of AD and considers OT as a promising therapeutic agent for addressing these early deficits.
Collapse
Affiliation(s)
- Daniela Černotová
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Karolína Hrůzová
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - David Levčík
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Svoboda
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Aleš Stuchlík
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
25
|
van den Berg M, Toen D, Verhoye M, Keliris GA. Alterations in theta-gamma coupling and sharp wave-ripple, signs of prodromal hippocampal network impairment in the TgF344-AD rat model. Front Aging Neurosci 2023; 15:1081058. [PMID: 37032829 PMCID: PMC10075364 DOI: 10.3389/fnagi.2023.1081058] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative disorder caused by the accumulation of toxic proteins, amyloid-beta (Aβ) and tau, which eventually leads to dementia. Disease-modifying therapies are still lacking, due to incomplete insights into the neuropathological mechanisms of AD. Synaptic dysfunction is known to occur before cognitive symptoms become apparent and recent studies have demonstrated that imbalanced synaptic signaling drives the progression of AD, suggesting that early synaptic dysfunction could be an interesting therapeutic target. Synaptic dysfunction results in altered oscillatory activity, which can be detected with electroencephalography and electrophysiological recordings. However, the majority of these studies have been performed at advanced stages of AD, when extensive damage and cognitive symptoms are already present. The current study aimed to investigate if the hippocampal oscillatory activity is altered at pre-plaque stages of AD. The rats received stereotactic surgery to implant a laminar electrode in the CA1 layer of the right hippocampus. Electrophysiological recordings during two consecutive days in an open field were performed in 4-5-month-old TgF344-AD rats when increased concentrations of soluble Aβ species were observed in the brain, in the absence of Aβ-plaques. We observed a decreased power of high theta oscillations in TgF344-AD rats compared to wild-type littermates. Sharp wave-ripple (SWR) analysis revealed an increased SWR power and a decreased duration of SWR during quiet wake in TgF344-AD rats. The alterations in properties of SWR and the increased power of fast oscillations are suggestive of neuronal hyperexcitability, as has been demonstrated to occur during presymptomatic stages of AD. In addition, decreased strength of theta-gamma coupling, an important neuronal correlate of memory encoding, was observed in the TgF344-AD rats. Theta-gamma phase amplitude coupling has been associated with memory encoding and the execution of cognitive functions. Studies have demonstrated that mild cognitive impairment patients display decreased coupling strength, similar to what is described here. The current study demonstrates altered hippocampal network activity occurring at pre-plaque stages of AD and provides insights into prodromal network dysfunction in AD. The alterations observed could aid in the detection of AD during presymptomatic stages.
Collapse
Affiliation(s)
- Monica van den Berg
- Bio-Imaging Lab, University of Antwerp, Wilrijk, Belgium
- μNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
- *Correspondence: Monica van den Berg, ; Georgios A. Keliris,
| | - Daniëlle Toen
- Bio-Imaging Lab, University of Antwerp, Wilrijk, Belgium
- μNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Marleen Verhoye
- Bio-Imaging Lab, University of Antwerp, Wilrijk, Belgium
- μNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Georgios A. Keliris
- Bio-Imaging Lab, University of Antwerp, Wilrijk, Belgium
- μNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Institute of Computer Science, Foundation for Research and Technology – Hellas, Heraklion, Crete, Greece
- *Correspondence: Monica van den Berg, ; Georgios A. Keliris,
| |
Collapse
|
26
|
Huang J, Wang M, Ju H, Shi Z, Ding W, Zhang D. SD-CNN: A static-dynamic convolutional neural network for functional brain networks. Med Image Anal 2023; 83:102679. [PMID: 36423466 DOI: 10.1016/j.media.2022.102679] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/14/2022] [Accepted: 10/29/2022] [Indexed: 11/13/2022]
Abstract
Static functional connections (sFCs) and dynamic functional connections (dFCs) have been widely used in the resting-state functional MRI (rs-fMRI) analysis. sFCs, calculated based on entire rs-fMRI scans, can accurately describe the static topology of the brain network. dFCs, estimated by dividing rs-fMRI scans into a series of short sliding windows, are used to reveal time-varying changes in FC patterns. Currently, how to jointly use sFCs and dFCs to identify brain diseases under the framework of deep learning is still a hot issue. To this end, we propose a static-dynamic convolutional neural network for functional brain networks, which involves a static pathway and a dynamic pathway for taking full advantages of sFCs and dFCs. Specifically, the static pathway, using high-resolution convolution filters (i.e., convolution filters with a high number of channels) at a single adjacency matrix of sFCs, is performed to capture static FC patterns. The dynamic pathway, using low-resolution convolution filters at each adjacency matrix of dFCs, is performed to capture time-varying FC patterns. Two types of diffusion connections are used in this model for encouraging the transfer of information between the static pathway and the dynamic pathway, which can make the learned features more discriminative. Furthermore, a static and dynamic combination classifier is introduced to combine features from two pathways for identifying brain diseases. Experiments on two real datasets demonstrate the effectiveness and advantages of our proposed method.
Collapse
Affiliation(s)
- Jiashuang Huang
- School of Information Science and Technology, Nantong University, Nantong, 226019, China; MIIT Key Laboratory of Pattern Analysis and Machine Intelligence, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Mingliang Wang
- School of Computer and Software, Nanjing University of Information Science and Technology, Nanjing, 210044, China; MIIT Key Laboratory of Pattern Analysis and Machine Intelligence, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Hengrong Ju
- School of Information Science and Technology, Nantong University, Nantong, 226019, China
| | - Zhenquan Shi
- School of Information Science and Technology, Nantong University, Nantong, 226019, China
| | - Weiping Ding
- School of Information Science and Technology, Nantong University, Nantong, 226019, China.
| | - Daoqiang Zhang
- Department of Computer Science and Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China.
| |
Collapse
|
27
|
Bonvicini G, Syvänen S, Andersson KG, Haaparanta-Solin M, López-Picón F, Sehlin D. ImmunoPET imaging of amyloid-beta in a rat model of Alzheimer's disease with a bispecific, brain-penetrating fusion protein. Transl Neurodegener 2022; 11:55. [PMID: 36567338 PMCID: PMC9791759 DOI: 10.1186/s40035-022-00324-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/03/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Hijacking the transferrin receptor (TfR) is an effective strategy to transport amyloid-beta (Aβ) immuno-positron emission tomography (immunoPET) ligands across the blood-brain barrier (BBB). Such ligands are more sensitive and specific than small-molecule ligands at detecting Aβ pathology in mouse models of Alzheimer's disease (AD). This study aimed to determine if this strategy would be as sensitive in rats and to assess how TfR affinity affects BBB transport of bispecific immunoPET radioligands. METHODS Two affinity variants of the rat TfR antibody, OX26, were chemically conjugated to a F(ab')2 fragment of the anti-Aβ antibody, bapineuzumab (Bapi), to generate two bispecific fusion proteins: OX265-F(ab')2-Bapi and OX2676-F(ab')2-Bapi. Pharmacokinetic analyses were performed 4 h and 70 h post-injection of radioiodinated fusion proteins in wild-type (WT) rats. [124I]I-OX265-F(ab')2-Bapi was administered to TgF344-AD and WT rats for in vivo PET imaging. Ex vivo distribution of injected [124I]I-OX265-F(ab')2-Bapi and Aβ pathology were assessed. RESULTS More [125I]I-OX265-F(ab')2-Bapi was taken up into the brain 4 h post-administration than [124I]I-OX2676-F(ab')2-Bapi. [124I]I-OX265-F(ab')2-Bapi PET visualized Aβ pathology with significantly higher signals in the TgF344-AD rats than in the WT littermates without Aβ pathology. The PET signals significantly correlated with Aβ levels in AD animals. CONCLUSION Affinity to TfR affects how efficiently a TfR-targeting bispecific fusion protein will cross the BBB, such that the higher-affinity bispecific fusion protein crossed the BBB more efficiently. Furthermore, bispecific immunoPET imaging of brain Aβ pathology using TfR-mediated transport provides good imaging contrast between TgF344-AD and WT rats, suggesting that this immunoPET strategy has the potential to be translated to higher species.
Collapse
Affiliation(s)
- Gillian Bonvicini
- grid.8993.b0000 0004 1936 9457Department of Public Health and Caring Sciences, Uppsala University, 751 85 Uppsala, Sweden ,BioArctic AB, 112 51 Stockholm, Sweden
| | - Stina Syvänen
- grid.8993.b0000 0004 1936 9457Department of Public Health and Caring Sciences, Uppsala University, 751 85 Uppsala, Sweden
| | | | - Merja Haaparanta-Solin
- grid.1374.10000 0001 2097 1371Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, 20520 Turku, Finland ,grid.1374.10000 0001 2097 1371MediCity Research Laboratory, University of Turku, 20520 Turku, Finland
| | - Francisco López-Picón
- grid.1374.10000 0001 2097 1371Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, 20520 Turku, Finland ,grid.1374.10000 0001 2097 1371MediCity Research Laboratory, University of Turku, 20520 Turku, Finland
| | - Dag Sehlin
- grid.8993.b0000 0004 1936 9457Department of Public Health and Caring Sciences, Uppsala University, 751 85 Uppsala, Sweden
| |
Collapse
|
28
|
Ceyzériat K, Zilli T, Millet P, Koutsouvelis N, Dipasquale G, Fossey C, Cailly T, Fabis F, Frisoni GB, Garibotto V, Tournier BB. Low-dose brain irradiation normalizes TSPO and CLUSTERIN levels and promotes the non-amyloidogenic pathway in pre-symptomatic TgF344-AD rats. J Neuroinflammation 2022; 19:311. [PMID: 36550510 PMCID: PMC9783748 DOI: 10.1186/s12974-022-02673-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Preclinical studies have recently evaluated the impact of low-dose brain radiation therapy (LD-RT) in animal models of Alzheimer's disease (AD) showing anti-amyloid and anti-inflammatory effects of this treatment. Its effectiveness varied, however, depending on the LD-RT protocol used and the stage when the treatment was applied. In this study, we aimed to evaluate the therapeutic potential of 10 Gy delivered in five daily fractions of 2 Gy (a protocol previously shown to induce an improvement of cognitive performances) in 9-month-old TgF344-AD rats, modeling at a pre-symptomatic stage of the disease. We showed that at an early stage, LD-RT was able to lower levels of the 18-kDa translocator protein (TSPO)-mediated neuroinflammation to normal ranges in addition to the secreted CLUSTERIN, another inflammatory protein also involved in Aβ aggregation. In addition, we demonstrated that LD-RT reduces all amyloid forms (~ - 60 to - 80%, P < 0.01; soluble and aggregated forms of Aβ40, Aβ42, and Aβoligomers). Interestingly, we showed for the first time that sAPPα levels were improved by the treatment, showing a higher activation of the non-amyloidogenic pathway, that could favor neuronal survival. The current evidence confirms the capacity of LD-RT to successfully modulate two pathological hallmarks of AD, namely amyloid and neuroinflammation, when applied before symptoms onset.
Collapse
Affiliation(s)
- Kelly Ceyzériat
- grid.8591.50000 0001 2322 4988Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, and Faculty of Medicine, Geneva University, Avenue de La Roseraie 64, 1205 Geneva, Switzerland ,grid.8591.50000 0001 2322 4988Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, Geneva University Hospitals, and NimtLab, Faculty of Medicine, Geneva University, 1205 Geneva, Switzerland ,grid.8591.50000 0001 2322 4988CIBM Center for BioMedical Imaging, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Thomas Zilli
- Department of Radiation Oncology, Oncology Institute of Southern Switzerland, EOC, 6500 Bellinzona, Switzerland ,grid.8591.50000 0001 2322 4988Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland ,grid.150338.c0000 0001 0721 9812Division of Radiation Oncology, Department of Oncology, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Philippe Millet
- grid.8591.50000 0001 2322 4988Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, and Faculty of Medicine, Geneva University, Avenue de La Roseraie 64, 1205 Geneva, Switzerland
| | - Nikolaos Koutsouvelis
- grid.150338.c0000 0001 0721 9812Division of Radiation Oncology, Department of Oncology, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Giovanna Dipasquale
- grid.150338.c0000 0001 0721 9812Division of Radiation Oncology, Department of Oncology, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Christine Fossey
- grid.412043.00000 0001 2186 4076Centre d’Études et de Recherche Sur le Médicament de Normandie (CERMN), Normandie Univ, UNICAEN, 1400 Caen, France
| | - Thomas Cailly
- grid.412043.00000 0001 2186 4076Centre d’Études et de Recherche Sur le Médicament de Normandie (CERMN), Normandie Univ, UNICAEN, 1400 Caen, France ,grid.411149.80000 0004 0472 0160Department of Nuclear Medicine, CHU Cote de Nacre, 1400 Caen, France ,grid.412043.00000 0001 2186 4076Normandie Univ, UNICAEN, IMOGERE, 1400 Caen, France ,Institut Blood and Brain @Caen-Normandie (BB@C), Boulevard Henri Becquerel, 14074 Caen, France
| | - Frédéric Fabis
- grid.412043.00000 0001 2186 4076Centre d’Études et de Recherche Sur le Médicament de Normandie (CERMN), Normandie Univ, UNICAEN, 1400 Caen, France
| | - Giovanni B. Frisoni
- grid.8591.50000 0001 2322 4988Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, Geneva University Hospitals, and NimtLab, Faculty of Medicine, Geneva University, 1205 Geneva, Switzerland
| | - Valentina Garibotto
- grid.8591.50000 0001 2322 4988Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, Geneva University Hospitals, and NimtLab, Faculty of Medicine, Geneva University, 1205 Geneva, Switzerland ,grid.8591.50000 0001 2322 4988CIBM Center for BioMedical Imaging, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Benjamin B. Tournier
- grid.8591.50000 0001 2322 4988Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, and Faculty of Medicine, Geneva University, Avenue de La Roseraie 64, 1205 Geneva, Switzerland
| |
Collapse
|
29
|
Measurement of Protein Synthesis Rate in Rat by [11C]Leucine PET Imaging: Application to the TgF344-AD Model of Alzheimer’s Disease. Mol Imaging Biol 2022; 25:596-605. [PMID: 36538180 PMCID: PMC10172255 DOI: 10.1007/s11307-022-01796-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/03/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022]
Abstract
AbstractLong-term memory requires stable protein synthesis and is altered in Alzheimer’s disease (AD). This study aimed to implement a method to measure the cerebral protein synthesis rate (PSR) with [11C]leucine PET in vivo in rats and evaluate potential PSR alterations longitudinally (6, 12 and 18 months old) in the TgF344-AD rat model of AD. Wistar, wild-type (WT) and TgF344-AD rats (TG) were scanned for 60 min with [11C]leucine. Arterial blood activity was monitored online and with discrete whole blood and plasma samples by γ-counting in Wistar rats, WT (n = 4) and TG (n = 5). Unlabelled amino acids were measured in plasma. The sensitivity of [11C]leucine PET to measure alterations in PSR was assessed in Wistar rats by injection of PSR inhibitor anisomycin before PET acquisition. Anisomycin administration significantly reduced the net uptake rate constant (Kcplx) of [11C]leucine and PSR, proving the suitability of the method. For the longitudinal study, averaged population-based input functions were used to calculate PSR. We found a significant genotype effect on PSR (decrease in TG vs WT) only in the globus pallidus. This study suggests that [11C]leucine PET is sensitive enough to measure brain PSR in rat but that cross-sectional design with individual input function should be preferred.
Collapse
|
30
|
Morisset C, Dizeux A, Larrat B, Selingue E, Boutin H, Picaud S, Sahel JA, Ialy-Radio N, Pezet S, Tanter M, Deffieux T. Retinal functional ultrasound imaging (rfUS) for assessing neurovascular alterations: a pilot study on a rat model of dementia. Sci Rep 2022; 12:19515. [PMID: 36376408 PMCID: PMC9663720 DOI: 10.1038/s41598-022-23366-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022] Open
Abstract
Fifty million people worldwide are affected by dementia, a heterogeneous neurodegenerative condition encompassing diseases such as Alzheimer's, vascular dementia, and Parkinson's. For them, cognitive decline is often the first marker of the pathology after irreversible brain damage has already occurred. Researchers now believe that structural and functional alterations of the brain vasculature could be early precursors of the diseases and are looking at how functional imaging could provide an early diagnosis years before irreversible clinical symptoms. In this preclinical pilot study, we proposed using functional ultrasound (fUS) on the retina to assess neurovascular alterations non-invasively, bypassing the skull limitation. We demonstrated for the first time the use of functional ultrasound in the retina and applied it to characterize the retinal hemodynamic response function in vivo in rats following a visual stimulus. We then demonstrated that retinal fUS could measure robust neurovascular coupling alterations between wild-type rats and TgF344-AD rat models of Alzheimer's disease. We observed an average relative increase in blood volume of 21% in the WT versus 37% for the TG group (p = 0.019). As a portable, non-invasive and inexpensive technique, rfUS is a promising functional screening tool in clinics for dementia years before symptoms.
Collapse
Affiliation(s)
- Clementine Morisset
- grid.440907.e0000 0004 1784 3645Institute Physics for Medicine Paris, INSERM U1273, ESPCI PSL Paris, CNRS UMR 8631, PSL Research University, Paris, France
| | - Alexandre Dizeux
- grid.440907.e0000 0004 1784 3645Institute Physics for Medicine Paris, INSERM U1273, ESPCI PSL Paris, CNRS UMR 8631, PSL Research University, Paris, France
| | - Benoit Larrat
- grid.457334.20000 0001 0667 2738NeuroSpin, Institut Des Sciences du Vivant Frédéric Joliot, Commissariat À L’Energie Atomique Et Aux Energies Alternatives (CEA), CNRS, Université Paris-Saclay, 91191 Gif-Sur-Yvette, France
| | - Erwan Selingue
- grid.457334.20000 0001 0667 2738NeuroSpin, Institut Des Sciences du Vivant Frédéric Joliot, Commissariat À L’Energie Atomique Et Aux Energies Alternatives (CEA), CNRS, Université Paris-Saclay, 91191 Gif-Sur-Yvette, France
| | - Herve Boutin
- grid.5379.80000000121662407Faculty of Biology, Medicine and Health, School of Biological Sciences Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, M13 9PL UK ,grid.5379.80000000121662407Wolfson Molecular Imaging Centre, University of Manchester, 27 Palatine Road, Manchester, M20 3LJ UK ,grid.462482.e0000 0004 0417 0074Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance and University of Manchester, Manchester, UK
| | - Serge Picaud
- grid.418241.a0000 0000 9373 1902Institut de La Vision, Sorbonne Université, INSERM, CNRS, 17 Rue Moreau, 75012 Paris, France
| | - Jose-Alain Sahel
- grid.418241.a0000 0000 9373 1902Institut de La Vision, Sorbonne Université, INSERM, CNRS, 17 Rue Moreau, 75012 Paris, France ,grid.21925.3d0000 0004 1936 9000Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA ,grid.417888.a0000 0001 2177 525XDepartment of Ophthalmology and Vitreo-Retinal Diseases, Fondation Ophtalmologique Rothschild, 75019 Paris, France
| | - Nathalie Ialy-Radio
- grid.440907.e0000 0004 1784 3645Institute Physics for Medicine Paris, INSERM U1273, ESPCI PSL Paris, CNRS UMR 8631, PSL Research University, Paris, France
| | - Sophie Pezet
- grid.440907.e0000 0004 1784 3645Institute Physics for Medicine Paris, INSERM U1273, ESPCI PSL Paris, CNRS UMR 8631, PSL Research University, Paris, France
| | - Mickael Tanter
- grid.440907.e0000 0004 1784 3645Institute Physics for Medicine Paris, INSERM U1273, ESPCI PSL Paris, CNRS UMR 8631, PSL Research University, Paris, France
| | - Thomas Deffieux
- grid.440907.e0000 0004 1784 3645Institute Physics for Medicine Paris, INSERM U1273, ESPCI PSL Paris, CNRS UMR 8631, PSL Research University, Paris, France
| |
Collapse
|
31
|
Lennol MP, Sánchez-Domínguez I, Cuchillo-Ibañez I, Camporesi E, Brinkmalm G, Alcolea D, Fortea J, Lleó A, Soria G, Aguado F, Zetterberg H, Blennow K, Sáez-Valero J. Apolipoprotein E imbalance in the cerebrospinal fluid of Alzheimer's disease patients. Alzheimers Res Ther 2022; 14:161. [PMID: 36324176 PMCID: PMC9628034 DOI: 10.1186/s13195-022-01108-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 10/16/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The purpose of this study was to examine the levels of cerebrospinal fluid (CSF) apolipoprotein E (apoE) species in Alzheimer's disease (AD) patients. METHODS We analyzed two CSF cohorts of AD and control individuals expressing different APOE genotypes. Moreover, CSF samples from the TgF344-AD rat model were included. Samples were run in native- and SDS-PAGE under reducing or non-reducing conditions (with or without β-mercaptoethanol). Immunoprecipitation combined with mass spectrometry or western blotting analyses served to assess the identity of apoE complexes. RESULTS In TgF344-AD rats expressing a unique apoE variant resembling human apoE4, a ~35-kDa apoE monomer was identified, increasing at 16.5 months compared with wild-types. In humans, apoE isoforms form disulfide-linked dimers in CSF, except apoE4, which lacks a cysteine residue. Thus, controls showed a decrease in the apoE dimer/monomer quotient in the APOE ε3/ε4 group compared with ε3/ε3 by native electrophoresis. A major contribution of dimers was found in APOE ε3/ε4 AD cases, and, unexpectedly, dimers were also found in ε4/ε4 AD cases. Under reducing conditions, two apoE monomeric glycoforms at 36 kDa and at 34 kDa were found in all human samples. In AD patients, the amount of the 34-kDa species increased, while the 36-kDa/34-kDa quotient was lower compared with controls. Interestingly, under reducing conditions, a ~100-kDa apoE complex, the identity of which was confirmed by mass spectrometry, also appeared in human AD individuals across all APOE genotypes, suggesting the occurrence of aberrantly resistant apoE aggregates. A second independent cohort of CSF samples validated these results. CONCLUSION These results indicate that despite the increase in total apoE content the apoE protein is altered in AD CSF, suggesting that function may be compromised.
Collapse
Affiliation(s)
- Matthew Paul Lennol
- grid.466805.90000 0004 1759 6875Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Av. Ramón y Cajal s/n, E-03550 Sant Joan d’Alacant, Spain ,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Sant Joan d’Alacant, Spain
| | - Irene Sánchez-Domínguez
- grid.5841.80000 0004 1937 0247Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain ,grid.5841.80000 0004 1937 0247Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Inmaculada Cuchillo-Ibañez
- grid.466805.90000 0004 1759 6875Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Av. Ramón y Cajal s/n, E-03550 Sant Joan d’Alacant, Spain ,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Sant Joan d’Alacant, Spain ,grid.513062.30000 0004 8516 8274Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Elena Camporesi
- grid.8761.80000 0000 9919 9582Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Gunnar Brinkmalm
- grid.8761.80000 0000 9919 9582Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Daniel Alcolea
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Sant Joan d’Alacant, Spain ,grid.7080.f0000 0001 2296 0625Sant Pau Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Juan Fortea
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Sant Joan d’Alacant, Spain ,grid.7080.f0000 0001 2296 0625Sant Pau Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain ,Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Alberto Lleó
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Sant Joan d’Alacant, Spain ,grid.7080.f0000 0001 2296 0625Sant Pau Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Guadalupe Soria
- grid.5841.80000 0004 1937 0247Institute of Neurosciences, University of Barcelona, Barcelona, Spain ,grid.5841.80000 0004 1937 0247Laboratory of Surgical Neuroanatomy, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Fernando Aguado
- grid.5841.80000 0004 1937 0247Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain ,grid.5841.80000 0004 1937 0247Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Henrik Zetterberg
- grid.8761.80000 0000 9919 9582Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden ,grid.1649.a000000009445082XClinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden ,grid.83440.3b0000000121901201Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK ,grid.83440.3b0000000121901201UK Dementia Research Institute at UCL, London, UK ,grid.24515.370000 0004 1937 1450Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- grid.8761.80000 0000 9919 9582Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden ,grid.1649.a000000009445082XClinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Javier Sáez-Valero
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Av. Ramón y Cajal s/n, E-03550, Sant Joan d'Alacant, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Sant Joan d'Alacant, Spain. .,Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain.
| |
Collapse
|
32
|
Spatio-temporal metabolic rewiring in the brain of TgF344-AD rat model of Alzheimer's disease. Sci Rep 2022; 12:16958. [PMID: 36216838 PMCID: PMC9550832 DOI: 10.1038/s41598-022-20962-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 09/21/2022] [Indexed: 12/29/2022] Open
Abstract
Brain damage associated with Alzheimer's disease (AD) occurs even decades before the symptomatic onset, raising the need to investigate its progression from prodromal stages. In this context, animal models that progressively display AD pathological hallmarks (e.g. TgF344-AD) become crucial. Translational technologies, such as magnetic resonance spectroscopy (MRS), enable the longitudinal metabolic characterization of this disease. However, an integrative approach is required to unravel the complex metabolic changes underlying AD progression, from early to advanced stages. TgF344-AD and wild-type (WT) rats were studied in vivo on a 7 Tesla MRI scanner, for longitudinal quantitative assessment of brain metabolic profile changes using MRS. Disease progression was investigated at 4 time points, from 9 to 18 months of age, and in 4 regions: cortex, hippocampus, striatum, and thalamus. Compared to WT, TgF344-AD rats replicated common findings in AD patients, including decreased N-acetylaspartate in the cortex, hippocampus and thalamus, and decreased glutamate in the thalamus and striatum. Different longitudinal evolution of metabolic concentration was observed between TgF344-AD and WT groups. Namely, age-dependent trajectories differed between groups for creatine in the cortex and thalamus and for taurine in cortex, with significant decreases in Tg344-AD animals; whereas myo-inositol in the thalamus and striatum showed greater increase along time in the WT group. Additional analysis revealed divergent intra- and inter-regional metabolic coupling in each group. Thus, in cortex, strong couplings of N-acetylaspartate and creatine with myo-inositol in WT, but with taurine in TgF344-AD rats were observed; whereas in the hippocampus, myo-inositol, taurine and choline compounds levels were highly correlated in WT but not in TgF344-AD animals. Furthermore, specific cortex-hippocampus-striatum metabolic crosstalks were found for taurine levels in the WT group but for myo-inositol levels in the TgF344-AD rats. With a systems biology perspective of metabolic changes in AD pathology, our results shed light into the complex spatio-temporal metabolic rewiring in this disease, reported here for the first time. Age- and tissue-dependent imbalances between myo-inositol, taurine and other metabolites, such as creatine, unveil their role in disease progression, while pointing to the inadequacy of the latter as an internal reference for quantification.
Collapse
|
33
|
van den Berg M, Adhikari MH, Verschuuren M, Pintelon I, Vasilkovska T, Van Audekerke J, Missault S, Heymans L, Ponsaerts P, De Vos WH, Van der Linden A, Keliris GA, Verhoye M. Altered basal forebrain function during whole-brain network activity at pre- and early-plaque stages of Alzheimer's disease in TgF344-AD rats. Alzheimers Res Ther 2022; 14:148. [PMID: 36217211 PMCID: PMC9549630 DOI: 10.1186/s13195-022-01089-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/22/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Imbalanced synaptic transmission appears to be an early driver in Alzheimer's disease (AD) leading to brain network alterations. Early detection of altered synaptic transmission and insight into mechanisms causing early synaptic alterations would be valuable treatment strategies. This study aimed to investigate how whole-brain networks are influenced at pre- and early-plague stages of AD and if these manifestations are associated with concomitant cellular and synaptic deficits. METHODS: To this end, we used an established AD rat model (TgF344-AD) and employed resting state functional MRI and quasi-periodic pattern (QPP) analysis, a method to detect recurrent spatiotemporal motifs of brain activity, in parallel with state-of-the-art immunohistochemistry in selected brain regions. RESULTS At the pre-plaque stage, QPPs in TgF344-AD rats showed decreased activity of the basal forebrain (BFB) and the default mode-like network. Histological analyses revealed increased astrocyte abundance restricted to the BFB, in the absence of amyloid plaques, tauopathy, and alterations in a number of cholinergic, gaba-ergic, and glutamatergic synapses. During the early-plaque stage, when mild amyloid-beta (Aβ) accumulation was observed in the cortex and hippocampus, QPPs in the TgF344-AD rats normalized suggesting the activation of compensatory mechanisms during this early disease progression period. Interestingly, astrogliosis observed in the BFB at the pre-plaque stage was absent at the early-plaque stage. Moreover, altered excitatory/inhibitory balance was observed in cortical regions belonging to the default mode-like network. In wild-type rats, at both time points, peak activity in the BFB preceded peak activity in other brain regions-indicating its modulatory role during QPPs. However, this pattern was eliminated in TgF344-AD suggesting that alterations in BFB-directed neuromodulation have a pronounced impact in network function in AD. CONCLUSIONS This study demonstrates the value of rsfMRI and advanced network analysis methods to detect early alterations in BFB function in AD, which could aid early diagnosis and intervention in AD. Restoring the global synaptic transmission, possibly by modulating astrogliosis in the BFB, might be a promising therapeutic strategy to restore brain network function and delay the onset of symptoms in AD.
Collapse
Affiliation(s)
- Monica van den Berg
- grid.5284.b0000 0001 0790 3681Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1 2610 Wilrijk, Antwerp, Belgium ,grid.5284.b0000 0001 0790 3681µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Mohit H. Adhikari
- grid.5284.b0000 0001 0790 3681Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1 2610 Wilrijk, Antwerp, Belgium ,grid.5284.b0000 0001 0790 3681µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Marlies Verschuuren
- grid.5284.b0000 0001 0790 3681µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium ,grid.5284.b0000 0001 0790 3681Laboratory of Cell Biology and Histology, University of Antwerp, Universiteitsplein 1 2610 Wilrijk, Antwerp, Belgium ,Antwerp Centre for Advanced Microscopy, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
| | - Isabel Pintelon
- grid.5284.b0000 0001 0790 3681µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium ,grid.5284.b0000 0001 0790 3681Laboratory of Cell Biology and Histology, University of Antwerp, Universiteitsplein 1 2610 Wilrijk, Antwerp, Belgium ,Antwerp Centre for Advanced Microscopy, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
| | - Tamara Vasilkovska
- grid.5284.b0000 0001 0790 3681Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1 2610 Wilrijk, Antwerp, Belgium ,grid.5284.b0000 0001 0790 3681µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Johan Van Audekerke
- grid.5284.b0000 0001 0790 3681Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1 2610 Wilrijk, Antwerp, Belgium ,grid.5284.b0000 0001 0790 3681µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Stephan Missault
- grid.5284.b0000 0001 0790 3681Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1 2610 Wilrijk, Antwerp, Belgium ,grid.5284.b0000 0001 0790 3681µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Loran Heymans
- grid.5284.b0000 0001 0790 3681Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1 2610 Wilrijk, Antwerp, Belgium ,grid.5284.b0000 0001 0790 3681µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Peter Ponsaerts
- grid.5284.b0000 0001 0790 3681Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
| | - Winnok H. De Vos
- grid.5284.b0000 0001 0790 3681µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium ,grid.5284.b0000 0001 0790 3681Laboratory of Cell Biology and Histology, University of Antwerp, Universiteitsplein 1 2610 Wilrijk, Antwerp, Belgium ,Antwerp Centre for Advanced Microscopy, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
| | - Annemie Van der Linden
- grid.5284.b0000 0001 0790 3681Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1 2610 Wilrijk, Antwerp, Belgium ,grid.5284.b0000 0001 0790 3681µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Georgios A. Keliris
- grid.5284.b0000 0001 0790 3681Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1 2610 Wilrijk, Antwerp, Belgium ,grid.5284.b0000 0001 0790 3681µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium ,grid.511960.aInstitute of Computer Science, Foundation for Research & Technology - Hellas, Heraklion, Crete, Greece
| | - Marleen Verhoye
- grid.5284.b0000 0001 0790 3681Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1 2610 Wilrijk, Antwerp, Belgium ,grid.5284.b0000 0001 0790 3681µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
34
|
Microglial NLRP3 inflammasome activates neurotoxic astrocytes in depression-like mice. Cell Rep 2022; 41:111532. [DOI: 10.1016/j.celrep.2022.111532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/22/2022] [Accepted: 09/29/2022] [Indexed: 11/17/2022] Open
|
35
|
Kimura T, Ono M, Seki C, Sampei K, Shimojo M, Kawamura K, Zhang MR, Sahara N, Takado Y, Higuchi M. A quantitative in vivo imaging platform for tracking pathological tau depositions and resultant neuronal death in a mouse model. Eur J Nucl Med Mol Imaging 2022; 49:4298-4311. [PMID: 35798978 DOI: 10.1007/s00259-022-05898-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 06/28/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE Depositions of tau fibrils are implicated in diverse neurodegenerative disorders, including Alzheimer's disease, and precise assessments of tau pathologies and their impacts on neuronal survival are crucial for pursuing the neurodegenerative tau pathogenesis with and without potential therapies. We aimed to establish an in vivo imaging system to quantify tau accumulations with positron emission tomography (PET) and brain atrophy with volumetric MRI in rTg4510 transgenic mice modeling neurodegenerative tauopathies. METHODS A total of 91 rTg4510 and non-transgenic control mice underwent PET with a tau radiotracer, 18F-PM-PBB3, and MRI at various ages (1.8-12.3 months). Using the cerebellum as reference, the radiotracer binding in target regions was estimated as standardized uptake value ratio (SUVR) and distribution volume ratio (DVR). Histopathological staining of brain sections derived from scanned animals was also conducted to investigate the imaging-neuropathology correlations. RESULTS 18F-PM-PBB3 SUVR at 40-60 min in the neocortex, hippocampus, and striatum of rTg4510 mice agreed with DVR, became significantly different from control values around 4-5 months of age, and progressively and negatively correlated with age and local volumes, respectively. Neocortical SUVR also correlated with the abundance of tau inclusions labeled with PM-PBB3 fluorescence, Gallyas-Braak silver impregnation, and anti-phospho-tau antibodies in postmortem assays. The in vivo and ex vivo 18F-PM-PBB3 binding was blocked by non-radioactive PM-PBB3. 18F-PM-PBB3 yielded a 1.6-fold greater dynamic range for tau imaging than its ancestor, 11C-PBB3. CONCLUSION Our imaging platform has enabled the quantification of tau depositions and consequent neuronal loss and is potentially applicable to the evaluation of candidate anti-tau and neuroprotective drugs.
Collapse
Affiliation(s)
- Taeko Kimura
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Maiko Ono
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Chie Seki
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan.
| | - Kazuaki Sampei
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Masafumi Shimojo
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Kazunori Kawamura
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Ming-Rong Zhang
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Naruhiko Sahara
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Yuhei Takado
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan.
| | - Makoto Higuchi
- National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| |
Collapse
|
36
|
Vagenknecht P, Luzgin A, Ono M, Ji B, Higuchi M, Noain D, Maschio CA, Sobek J, Chen Z, Konietzko U, Gerez JA, Riek R, Razansky D, Klohs J, Nitsch RM, Dean-Ben XL, Ni R. Non-invasive imaging of tau-targeted probe uptake by whole brain multi-spectral optoacoustic tomography. Eur J Nucl Med Mol Imaging 2022; 49:2137-2152. [PMID: 35128565 PMCID: PMC9165274 DOI: 10.1007/s00259-022-05708-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/25/2022] [Indexed: 02/06/2023]
Abstract
PURPOSE Abnormal tau accumulation within the brain plays an important role in tauopathies such as Alzheimer's disease and frontotemporal dementia. High-resolution imaging of tau deposits at the whole-brain scale in animal disease models is highly desired. METHODS We approached this challenge by non-invasively imaging the brains of P301L mice of 4-repeat tau with concurrent volumetric multi-spectral optoacoustic tomography (vMSOT) at ~ 115 μm spatial resolution using the tau-targeted pyridinyl-butadienyl-benzothiazole derivative PBB5 (i.v.). In vitro probe characterization, concurrent vMSOT and epi-fluorescence imaging of in vivo PBB5 targeting (i.v.) was performed in P301L and wild-type mice, followed by ex vivo validation using AT-8 antibody for phosphorylated tau. RESULTS PBB5 showed specific binding to recombinant K18 tau fibrils by fluorescence assay, to post-mortem Alzheimer's disease brain tissue homogenate by competitive binding against [11C]PBB3 and to tau deposits (AT-8 positive) in post-mortem corticobasal degeneration and progressive supranuclear palsy brains. Dose-dependent optoacoustic and fluorescence signal intensities were observed in the mouse brains following i.v. administration of different concentrations of PBB5. In vivo vMSOT brain imaging of P301L mice showed higher retention of PBB5 in the tau-laden cortex and hippocampus compared to wild-type mice, as confirmed by ex vivo vMSOT, epi-fluorescence, multiphoton microscopy, and immunofluorescence staining. CONCLUSIONS We demonstrated non-invasive whole-brain imaging of tau in P301L mice with vMSOT system using PBB5 at a previously unachieved ~ 115 μm spatial resolution. This platform provides a new tool to study tau spreading and clearance in a tauopathy mouse model, foreseeable in monitoring tau targeting putative therapeutics.
Collapse
Affiliation(s)
- Patrick Vagenknecht
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Artur Luzgin
- Zentrum für Neurowissenschaften Zürich (ZNZ), Zurich, Switzerland
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, ETH Zurich & University of Zurich, Zurich, Switzerland
| | - Maiko Ono
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Bin Ji
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- Department of Radiopharmacy and Molecular Imaging, School of Pharmacy, Fudan University, Shanghai, China
| | - Makoto Higuchi
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Daniela Noain
- Neurology Department, University Hospital Zurich, Zurich, Switzerland
| | - Cinzia A Maschio
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Zentrum für Neurowissenschaften Zürich (ZNZ), Zurich, Switzerland
| | - Jens Sobek
- Functional Genomics Center, University of Zurich, Zurich, Switzerland
| | - Zhenyue Chen
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, ETH Zurich & University of Zurich, Zurich, Switzerland
| | - Uwe Konietzko
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Juan A Gerez
- Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Roland Riek
- Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Daniel Razansky
- Zentrum für Neurowissenschaften Zürich (ZNZ), Zurich, Switzerland
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, ETH Zurich & University of Zurich, Zurich, Switzerland
| | - Jan Klohs
- Zentrum für Neurowissenschaften Zürich (ZNZ), Zurich, Switzerland
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, ETH Zurich & University of Zurich, Zurich, Switzerland
| | - Roger M Nitsch
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Zentrum für Neurowissenschaften Zürich (ZNZ), Zurich, Switzerland
| | - Xose Luis Dean-Ben
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, ETH Zurich & University of Zurich, Zurich, Switzerland.
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.
- Zentrum für Neurowissenschaften Zürich (ZNZ), Zurich, Switzerland.
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, ETH Zurich & University of Zurich, Zurich, Switzerland.
| |
Collapse
|
37
|
Chen B, Marquez-Nostra B, Belitzky E, Toyonaga T, Tong J, Huang Y, Cai Z. PET Imaging in Animal Models of Alzheimer’s Disease. Front Neurosci 2022; 16:872509. [PMID: 35685772 PMCID: PMC9171374 DOI: 10.3389/fnins.2022.872509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
The successful development and translation of PET imaging agents targeting β-amyloid plaques and hyperphosphorylated tau tangles have allowed for in vivo detection of these hallmarks of Alzheimer’s disease (AD) antemortem. Amyloid and tau PET have been incorporated into the A/T/N scheme for AD characterization and have become an integral part of ongoing clinical trials to screen patients for enrollment, prove drug action mechanisms, and monitor therapeutic effects. Meanwhile, preclinical PET imaging in animal models of AD can provide supportive information for mechanistic studies. With the recent advancement of gene editing technologies and AD animal model development, preclinical PET imaging in AD models will further facilitate our understanding of AD pathogenesis/progression and the development of novel treatments. In this study, we review the current state-of-the-art in preclinical PET imaging using animal models of AD and suggest future research directions.
Collapse
|
38
|
Syvänen S, Meier SR, Roshanbin S, Xiong M, Faresjö R, Gustavsson T, Bonvicini G, Schlein E, Aguilar X, Julku U, Eriksson J, Sehlin D. PET Imaging in Preclinical Anti-Aβ Drug Development. Pharm Res 2022; 39:1481-1496. [PMID: 35501533 PMCID: PMC9246809 DOI: 10.1007/s11095-022-03277-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/25/2022] [Indexed: 11/21/2022]
Abstract
Positron emission tomography (PET), a medical imaging technique allowing for studies of the living human brain, has gained an important role in clinical trials of novel drugs against Alzheimer’s disease (AD). For example, PET data contributed to the conditional approval in 2021 of aducanumab, an antibody directed towards amyloid-beta (Aβ) aggregates, by showing a dose-dependent reduction in brain amyloid after treatment. In parallel to clinical studies, preclinical studies in animal models of Aβ pathology may also benefit from PET as a tool to detect target engagement and treatment effects of anti-Aβ drug candidates. PET is associated with a high level of translatability between species as similar, non-invasive protocols allow for longitudinal rather than cross-sectional studies and can be used both in a preclinical and clinical setting. This review focuses on the use of preclinical PET imaging in genetically modified animals that express human Aβ, and its present and potential future role in the development of drugs aimed at reducing brain Aβ levels as a therapeutic strategy to halt disease progression in AD.
Collapse
Affiliation(s)
- Stina Syvänen
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden.
| | - Silvio R Meier
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Sahar Roshanbin
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Mengfei Xiong
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Rebecca Faresjö
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Tobias Gustavsson
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Gillian Bonvicini
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden.,BioArctic AB, Stockholm, Sweden
| | - Eva Schlein
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Ximena Aguilar
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Ulrika Julku
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Jonas Eriksson
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden.,PET Centre, Uppsala University Hospital, Uppsala, Sweden
| | - Dag Sehlin
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| |
Collapse
|
39
|
Nillert N, Boonyarat C, Welbat JU, Bunreungthong K, Puthongking P, Pannangrong W. Clausena Harmandiana root extract attenuated cognitive impairments via reducing amyloid accumulation and neuroinflammation in Aβ 1-42-induced rats. BMC Complement Med Ther 2022; 22:108. [PMID: 35439990 PMCID: PMC9019931 DOI: 10.1186/s12906-022-03591-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/11/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) pathogenesis is associated with amyloid-β (Aβ)-induced neuroinflammation. In AD, the activation of microglia caused by Aβ accumulation is followed by the synthesis and release of pro-inflammatory cytokines, including interleukin-1β (IL-1β) and tumor necrosis factor-α (TNFα), and ultimately leads to cognitive impairments. Clausena harmandiana (CH) is a medicinal plant in the Rutaceae family and has been used in folk medicine to relieve illnesses such as stomachache and headache, and as a health tonic. Interestingly, CH root extract (CHRE) has several anti-inflammatory and other pharmacological activities, but there are no studies in AD-like animal models. OBJECTIVES This study aims to evaluate the effects of CHRE on cognitive impairments, increased Aβ1-42 protein levels, and neuroinflammation in Aβ1-42-induced rats. METHODS Forty-eight adult male Sprague-Dawley rats (250-300 g) were randomly divided into 6 groups (n = 8) of the sham control, V + Aβ, CB + Aβ CHRE125 + Aβ, CHRE250 + Aβ, and CHRE500 + Aβ. Sodium carboxymethylcellulose, Celebrex (10 mg/kg BW) and CHRE (125, 250, and 500 mg/kg BW) were given orally or without any treatment for 35 days. On day 21, aggregated Aβ1-42 at a concentration of 1 μg/μl were injected into both lateral ventricles (1 μl/side) of all treated rats, while sterilized normal saline were injected to untreated rats. Ten days later, the novel object recognition test was performed to assess their recognition memory. At the end of the test period, an overdose of thiopental sodium (120 mg/kg BW) and transcardial perfusion with 0.9% normal saline solution were used to euthanize all rats. Then Aβ1-42 protein levels and the expression of inflammatory markers (CD11b-positive microglia, IL-1β, and TNFα) were investigated in the cerebral cortex and hippocampus. RESULTS Pretreatment with CHRE at all doses could attenuate short- and long-term impairments in recognition memory. Additionally, CHRE also inhibited the increase of Aβ1-42 protein levels and the expression of inflammatory markers in both brain regions as well as receiving Celebrex. CONCLUSIONS This suggests that preventive treatment of CHRE might be a potential therapy against cognitive impairments via reducing Aβ1-42 protein levels and neuroinflammation caused by Aβ1-42.
Collapse
Affiliation(s)
- Nutchareeporn Nillert
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Chantana Boonyarat
- Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Jariya Umka Welbat
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Komsun Bunreungthong
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Ploenthip Puthongking
- Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Wanassanun Pannangrong
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.
| |
Collapse
|
40
|
Shimochi S, Keller T, Kujala E, Khabbal J, Rajander J, Löyttyniemi E, Solin O, Nuutila P, Kanaya S, Yatkin E, Grönroos TJ, Iida H. Evaluation of [ 18F]F-DPA PET for Detecting Microglial Activation in the Spinal Cord of a Rat Model of Neuropathic Pain. Mol Imaging Biol 2022; 24:641-650. [PMID: 35303205 PMCID: PMC9296394 DOI: 10.1007/s11307-022-01713-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/15/2022] [Accepted: 02/18/2022] [Indexed: 12/05/2022]
Abstract
Purpose Recent studies have linked activated spinal glia to neuropathic pain. Here, using a positron emission tomography (PET) scanner with high spatial resolution and sensitivity, we evaluated the feasibility and sensitivity of N,N-diethyl-2-(2-(4-([18F]fluoro)phenyl)-5,7-dimethylpyrazolo[1,5-a] pyrimidin-3-yl)acetamide ([18F]F-DPA) imaging for detecting spinal cord microglial activation after partial sciatic nerve ligation (PSNL) in rats. Procedures Neuropathic pain was induced in rats (n = 20) by PSNL, and pain sensation tests were conducted before surgery and 3 and 7 days post-injury. On day 7, in vivo PET imaging and ex vivo autoradiography were performed using [18F]F-DPA or [11C]PK11195. Ex vivo biodistribution and PET imaging of the removed spinal cord were carried out with [18F]F-DPA. Sham-operated and PK11195-pretreated animals were also examined. Results Mechanical allodynia was confirmed in the PSNL rats from day 3 through day 7. Ex vivo autoradiography showed a higher lesion-to-background uptake with [18F]F-DPA compared with [11C]PK11195. Ex vivo PET imaging of the removed spinal cord showed [18F]F-DPA accumulation in the inflammation site, which was immunohistochemically confirmed to coincide with microglia activation. Pretreatment with PK11195 eliminated the uptake. The SUV values of in vivo [18F]F-DPA and [11C]PK11195 PET were not significantly increased in the lesion compared with the reference region, and were fivefold higher than the values obtained from the ex vivo data. Ex vivo biodistribution revealed a twofold higher [18F]F-DPA uptake in the vertebral body compared to that seen in the bone from the skull. Conclusions [18F]F-DPA aided visualization of the spinal cord inflammation site in PSNL rats on ex vivo autoradiography and was superior to [11C]PK11195. In vivo [18F]F-DPA PET did not allow for visualization of tracer accumulation even using a high-spatial-resolution PET scanner. The main reason for this result was due to insufficient SUVs in the spinal cord region as compared with the background noise, in addition to a spillover from the vertebral body.
Collapse
Affiliation(s)
- Saeka Shimochi
- Turku PET Centre, University of Turku, Turku, Finland.,Medicity Research Laboratory, University of Turku, Turku, Finland.,Nara Institute of Science and Technology, Ikoma City, Japan
| | - Thomas Keller
- Turku PET Centre, University of Turku, Turku, Finland
| | - Ella Kujala
- Central Animal Laboratory, University of Turku, Turku, Finland
| | - Joonas Khabbal
- Central Animal Laboratory, University of Turku, Turku, Finland
| | - Johan Rajander
- Accelerator Laboratory, Turku PET Centre, Åbo Akademi University, Turku, Finland
| | | | - Olof Solin
- Turku PET Centre, University of Turku, Turku, Finland.,Accelerator Laboratory, Turku PET Centre, Åbo Akademi University, Turku, Finland
| | - Pirjo Nuutila
- Turku PET Centre, University of Turku, Turku, Finland
| | | | - Emrah Yatkin
- Central Animal Laboratory, University of Turku, Turku, Finland
| | - Tove J Grönroos
- Turku PET Centre, University of Turku, Turku, Finland.,Medicity Research Laboratory, University of Turku, Turku, Finland
| | - Hidehiro Iida
- Turku PET Centre, University of Turku, Turku, Finland. .,Nara Institute of Science and Technology, Ikoma City, Japan. .,Turku PET Centre, Turku University Hospital, Turku, Finland.
| |
Collapse
|
41
|
Rafiee Z, García-Serrano AM, Duarte JMN. Taurine Supplementation as a Neuroprotective Strategy upon Brain Dysfunction in Metabolic Syndrome and Diabetes. Nutrients 2022; 14:1292. [PMID: 35334949 PMCID: PMC8952284 DOI: 10.3390/nu14061292] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 02/07/2023] Open
Abstract
Obesity, type 2 diabetes, and their associated comorbidities impact brain metabolism and function and constitute risk factors for cognitive impairment. Alterations to taurine homeostasis can impact a number of biological processes, such as osmolarity control, calcium homeostasis, and inhibitory neurotransmission, and have been reported in both metabolic and neurodegenerative disorders. Models of neurodegenerative disorders show reduced brain taurine concentrations. On the other hand, models of insulin-dependent diabetes, insulin resistance, and diet-induced obesity display taurine accumulation in the hippocampus. Given the possible cytoprotective actions of taurine, such cerebral accumulation of taurine might constitute a compensatory mechanism that attempts to prevent neurodegeneration. The present article provides an overview of brain taurine homeostasis and reviews the mechanisms by which taurine can afford neuroprotection in individuals with obesity and diabetes. We conclude that further research is needed for understanding taurine homeostasis in metabolic disorders with an impact on brain function.
Collapse
Affiliation(s)
- Zeinab Rafiee
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22100 Lund, Sweden; (Z.R.); (A.M.G.-S.)
- Wallenberg Centre for Molecular Medicine, Lund University, 22100 Lund, Sweden
| | - Alba M. García-Serrano
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22100 Lund, Sweden; (Z.R.); (A.M.G.-S.)
- Wallenberg Centre for Molecular Medicine, Lund University, 22100 Lund, Sweden
| | - João M. N. Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22100 Lund, Sweden; (Z.R.); (A.M.G.-S.)
- Wallenberg Centre for Molecular Medicine, Lund University, 22100 Lund, Sweden
| |
Collapse
|
42
|
Ni R. Magnetic Resonance Imaging in Tauopathy Animal Models. Front Aging Neurosci 2022; 13:791679. [PMID: 35145392 PMCID: PMC8821905 DOI: 10.3389/fnagi.2021.791679] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/27/2021] [Indexed: 11/13/2022] Open
Abstract
The microtubule-associated protein tau plays an important role in tauopathic diseases such as Alzheimer's disease and primary tauopathies such as progressive supranuclear palsy and corticobasal degeneration. Tauopathy animal models, such as transgenic, knock-in mouse and rat models, recapitulating tauopathy have facilitated the understanding of disease mechanisms. Aberrant accumulation of hyperphosphorylated tau contributes to synaptic deficits, neuroinflammation, and neurodegeneration, leading to cognitive impairment in animal models. Recent advances in molecular imaging using positron emission tomography (PET) and magnetic resonance imaging (MRI) have provided valuable insights into the time course of disease pathophysiology in tauopathy animal models. High-field MRI has been applied for in vivo imaging in animal models of tauopathy, including diffusion tensor imaging for white matter integrity, arterial spin labeling for cerebral blood flow, resting-state functional MRI for functional connectivity, volumetric MRI for neurodegeneration, and MR spectroscopy. In addition, MR contrast agents for non-invasive imaging of tau have been developed recently. Many preclinical MRI indicators offer excellent translational value and provide a blueprint for clinical MRI in the brains of patients with tauopathies. In this review, we summarized the recent advances in using MRI to visualize the pathophysiology of tauopathy in small animals. We discussed the outstanding challenges in brain imaging using MRI in small animals and propose a future outlook for visualizing tau-related alterations in the brains of animal models.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, ETH Zurich and University of Zurich, Zurich, Switzerland
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
43
|
Cao L, Kong Y, Ji B, Ren Y, Guan Y, Ni R. Positron Emission Tomography in Animal Models of Tauopathies. Front Aging Neurosci 2022; 13:761913. [PMID: 35082657 PMCID: PMC8784812 DOI: 10.3389/fnagi.2021.761913] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/30/2021] [Indexed: 12/18/2022] Open
Abstract
The microtubule-associated protein tau (MAPT) plays an important role in Alzheimer's disease and primary tauopathy diseases. The abnormal accumulation of tau contributes to the development of neurotoxicity, inflammation, neurodegeneration, and cognitive deficits in tauopathy diseases. Tau synergically interacts with amyloid-beta in Alzheimer's disease leading to detrimental consequence. Thus, tau has been an important target for therapeutics development for Alzheimer's disease and primary tauopathy diseases. Tauopathy animal models recapitulating the tauopathy such as transgenic, knock-in mouse and rat models have been developed and greatly facilitated the understanding of disease mechanisms. The advance in PET and imaging tracers have enabled non-invasive detection of the accumulation and spread of tau, the associated microglia activation, metabolic, and neurotransmitter receptor alterations in disease animal models. In vivo microPET studies on mouse or rat models of tauopathy have provided significant insights into the phenotypes and time course of pathophysiology of these models and allowed the monitoring of treatment targeting at tau. In this study, we discuss the utilities of PET and recently developed tracers for evaluating the pathophysiology in tauopathy animal models. We point out the outstanding challenges and propose future outlook in visualizing tau-related pathophysiological changes in brain of tauopathy disease animal models.
Collapse
Affiliation(s)
- Lei Cao
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Changes Technology Corporation Ltd., Shanghai, China
| | - Yanyan Kong
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Bin Ji
- Department of Radiopharmacy and Molecular Imaging, School of Pharmacy, Fudan University, Shanghai, China
| | - Yutong Ren
- Guangdong Robotics Association, Guangzhou, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, ETH Zurich and University of Zurich, Zurich, Switzerland
| |
Collapse
|
44
|
Fowler CF, Goerzen D, Devenyi GA, Madularu D, Chakravarty MM, Near J. OUP accepted manuscript. Brain Commun 2022; 4:fcac072. [PMID: 35434622 PMCID: PMC9007326 DOI: 10.1093/braincomms/fcac072] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/12/2022] [Accepted: 03/16/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Caitlin F. Fowler
- Department of Biological and Biomedical Engineering, McGill University, Duff Medical Building, Montreal, Canada H3A 2B4
- Centre d’Imagerie Cérébrale, Douglas Mental Health University Institute, Verdun, Canada H4H 1R3
- Correspondence to: Caitlin F. Fowler, CIC Pavilion Office GH-2113 Douglas Mental Health University Institute 6875 Boulevard LaSalle Montreal, Canada H4H 1R3 E-mail:
| | - Dana Goerzen
- Centre d’Imagerie Cérébrale, Douglas Mental Health University Institute, Verdun, Canada H4H 1R3
| | - Gabriel A. Devenyi
- Centre d’Imagerie Cérébrale, Douglas Mental Health University Institute, Verdun, Canada H4H 1R3
- Department of Psychiatry, McGill University, Montreal, Canada H3A 1A1
| | - Dan Madularu
- Centre for Translational NeuroImaging, Northeastern University, Boston, USA
| | - M. Mallar Chakravarty
- Department of Biological and Biomedical Engineering, McGill University, Duff Medical Building, Montreal, Canada H3A 2B4
- Centre d’Imagerie Cérébrale, Douglas Mental Health University Institute, Verdun, Canada H4H 1R3
- Department of Psychiatry, McGill University, Montreal, Canada H3A 1A1
| | - Jamie Near
- Department of Biological and Biomedical Engineering, McGill University, Duff Medical Building, Montreal, Canada H3A 2B4
- Centre d’Imagerie Cérébrale, Douglas Mental Health University Institute, Verdun, Canada H4H 1R3
- Department of Psychiatry, McGill University, Montreal, Canada H3A 1A1
- Physical Studies Research Platform, Sunnybrook Research Institute, Toronto, Canada M4N 3M5
- Department of Medical Biophysics, University of Toronto, Toronto, Canada M5G 1L7
| |
Collapse
|
45
|
Ni R. Magnetic Resonance Imaging in Animal Models of Alzheimer's Disease Amyloidosis. Int J Mol Sci 2021; 22:12768. [PMID: 34884573 PMCID: PMC8657987 DOI: 10.3390/ijms222312768] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
Amyloid-beta (Aβ) plays an important role in the pathogenesis of Alzheimer's disease. Aberrant Aβ accumulation induces neuroinflammation, cerebrovascular alterations, and synaptic deficits, leading to cognitive impairment. Animal models recapitulating the Aβ pathology, such as transgenic, knock-in mouse and rat models, have facilitated the understanding of disease mechanisms and the development of therapeutics targeting Aβ. There is a rapid advance in high-field MRI in small animals. Versatile high-field magnetic resonance imaging (MRI) sequences, such as diffusion tensor imaging, arterial spin labeling, resting-state functional MRI, anatomical MRI, and MR spectroscopy, as well as contrast agents, have been developed for preclinical imaging in animal models. These tools have enabled high-resolution in vivo structural, functional, and molecular readouts with a whole-brain field of view. MRI has been used to visualize non-invasively the Aβ deposits, synaptic deficits, regional brain atrophy, impairment in white matter integrity, functional connectivity, and cerebrovascular and glymphatic system in animal models of Alzheimer's disease amyloidosis. Many of the readouts are translational toward clinical MRI applications in patients with Alzheimer's disease. In this review, we summarize the recent advances in MRI for visualizing the pathophysiology in amyloidosis animal models. We discuss the outstanding challenges in brain imaging using MRI in small animals and propose future outlook in visualizing Aβ-related alterations in the brains of animal models.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, ETH Zurich & University of Zurich, 8093 Zurich, Switzerland;
- Institute for Regenerative Medicine, University of Zurich, 8952 Zurich, Switzerland
| |
Collapse
|
46
|
Ni R. Positron Emission Tomography in Animal Models of Alzheimer's Disease Amyloidosis: Translational Implications. Pharmaceuticals (Basel) 2021; 14:1179. [PMID: 34832961 PMCID: PMC8623863 DOI: 10.3390/ph14111179] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/13/2021] [Accepted: 11/15/2021] [Indexed: 12/30/2022] Open
Abstract
Animal models of Alzheimer's disease amyloidosis that recapitulate cerebral amyloid-beta pathology have been widely used in preclinical research and have greatly enabled the mechanistic understanding of Alzheimer's disease and the development of therapeutics. Comprehensive deep phenotyping of the pathophysiological and biochemical features in these animal models is essential. Recent advances in positron emission tomography have allowed the non-invasive visualization of the alterations in the brain of animal models and in patients with Alzheimer's disease. These tools have facilitated our understanding of disease mechanisms and provided longitudinal monitoring of treatment effects in animal models of Alzheimer's disease amyloidosis. In this review, we focus on recent positron emission tomography studies of cerebral amyloid-beta accumulation, hypoglucose metabolism, synaptic and neurotransmitter receptor deficits (cholinergic and glutamatergic system), blood-brain barrier impairment, and neuroinflammation (microgliosis and astrocytosis) in animal models of Alzheimer's disease amyloidosis. We further propose the emerging targets and tracers for reflecting the pathophysiological changes and discuss outstanding challenges in disease animal models and future outlook in the on-chip characterization of imaging biomarkers towards clinical translation.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, ETH & University of Zurich, 8093 Zurich, Switzerland;
- Institute for Regenerative Medicine, University of Zurich, 8952 Zurich, Switzerland
| |
Collapse
|
47
|
Filip T, Mairinger S, Neddens J, Sauberer M, Flunkert S, Stanek J, Wanek T, Okamura N, Langer O, Hutter-Paier B, Kuntner C. Characterization of an APP/tau rat model of Alzheimer's disease by positron emission tomography and immunofluorescent labeling. Alzheimers Res Ther 2021; 13:175. [PMID: 34656177 PMCID: PMC8522096 DOI: 10.1186/s13195-021-00916-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 10/05/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND To better understand the etiology and pathomechanisms of Alzheimer's disease, several transgenic animal models that overexpress human tau or human amyloid-beta (Aβ) have been developed. In the present study, we generated a novel transgenic rat model by cross-breeding amyloid precursor protein (APP) rats with tau rats. We characterized this model by performing positron emission tomography scans combined with immunofluorescent labeling and cerebrospinal fluid analyses. METHODS APP/Tau rats were generated by cross-breeding male McGill-R-Thy1-APP transgenic rats with female hTau-40/P301L transgenic rats. APP/Tau double transgenic rats and non-transgenic (ntg) littermates aged 7, 13, and 21 months were subjected to dynamic [11C] PiB scan and dynamic [18F]THK-5317 scans. For regional brain analysis, a template was generated from anatomical MR images of selected animals, which was co-registered with the PET images. Regional analysis was performed by application of the simplified reference tissue model ([11C]PiB data), whereas [18F]THK-5317 data were analyzed using a 2-tissue compartment model and Logan graphical analysis. In addition, immunofluorescent labeling (tau, amyloid) and cerebrospinal fluid analyses were performed. RESULTS [11C]PiB binding potential (BPND) and [18F]THK-5317 volume of distribution (VT) showed an increase with age in several brain regions in the APP/Tau group but not in the ntg control group. Immunohistochemical analysis of brain slices of PET-scanned animals revealed a positive correlation between Aβ labeling and [11C]PiB regional BPND. Tau staining yielded a trend towards higher levels in the cortex and hippocampus of APP/Tau rats compared with ntg littermates, but without reaching statistical significance. No correlation was found between tau immunofluorescence labeling results and the respective [18F]THK-5317 VT values. CONCLUSIONS We thoroughly characterized a novel APP/Tau rat model using combined PET imaging and immunofluorescence analysis. We observed an age-related increase in [11C]PiB and [18F]THK-5317 binding in several brain regions in the APP/Tau group but not in the ntg group. Although we were able to reveal a positive correlation between amyloid labeling and [11C]PiB regional brain uptake, we observed relatively low human tau and amyloid fibril expression levels and a somewhat unstable brain pathology which questions the utility of this animal model for further studies.
Collapse
Affiliation(s)
- Thomas Filip
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria
- Department of Biomedical Research, Medical University Vienna, Vienna, Austria
| | - Severin Mairinger
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Joerg Neddens
- Neuropharmacology, QPS Austria GmbH, Grambach, Austria
| | - Michael Sauberer
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | | | - Johann Stanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Thomas Wanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Oliver Langer
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | | | - Claudia Kuntner
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria.
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
48
|
Direct Comparison of [ 18F]F-DPA with [ 18F]DPA-714 and [ 11C]PBR28 for Neuroinflammation Imaging in the same Alzheimer's Disease Model Mice and Healthy Controls. Mol Imaging Biol 2021; 24:157-166. [PMID: 34542805 PMCID: PMC8760190 DOI: 10.1007/s11307-021-01646-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/22/2021] [Accepted: 08/23/2021] [Indexed: 11/02/2022]
Abstract
PURPOSE In this study we compared the recently developed TSPO tracer [18F]F-DPA, with [18F]DPA-714 and [11C]PBR28 by performing in vivo PET imaging on the same Alzheimer's disease mouse model APP/PS1-21 (TG) and wild-type (WT) mice with all three radiotracers. PROCEDURES To compare the radiotracer uptake, percentage of injected dose/mL (%ID/mL), standardized uptake value ratios to cerebellum (SUVRCB), and voxel-wise analyses were performed. RESULTS The peak uptake of [18F]F-DPA was higher than 4.3% ID/mL, while [18F]DPA-714 reached just over 3% ID/mL, and [11C]PBR28 was over 4% ID/mL in only one brain region in the WT mice. The peak/60-min uptake ratios of [18F]F-DPA were significantly higher (p < 0.001) than those of [18F]DPA-714 and [11C]PBR28. The differences in [18F]F-DPA SUVRCB between WT and TG mice were highly significant (p < 0.001) in the three studied time periods after injection. [18F]DPA-714 uptake was significantly higher in TG mice starting in the 20-40-min timeframe and increased thereafter, whereas [11C]PBR28 uptake became significant at 10-20 min (p < 0.05). The voxel-wise analysis confirmed the differences between the radiotracers. CONCLUSIONS [18F]F-DPA displays higher brain uptake, higher TG-to-WT SUVRCB ratios, and faster clearance than [18F]DPA-714 and [11C]PBR28, and could prove useful for detecting low levels of inflammation and allow for shorter dynamic PET scans.
Collapse
|
49
|
Zhou R, Ji B, Kong Y, Qin L, Ren W, Guan Y, Ni R. PET Imaging of Neuroinflammation in Alzheimer's Disease. Front Immunol 2021; 12:739130. [PMID: 34603323 PMCID: PMC8481830 DOI: 10.3389/fimmu.2021.739130] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 08/27/2021] [Indexed: 12/15/2022] Open
Abstract
Neuroinflammation play an important role in Alzheimer's disease pathogenesis. Advances in molecular imaging using positron emission tomography have provided insights into the time course of neuroinflammation and its relation with Alzheimer's disease central pathologies in patients and in animal disease models. Recent single-cell sequencing and transcriptomics indicate dynamic disease-associated microglia and astrocyte profiles in Alzheimer's disease. Mitochondrial 18-kDa translocator protein is the most widely investigated target for neuroinflammation imaging. New generation of translocator protein tracers with improved performance have been developed and evaluated along with tau and amyloid imaging for assessing the disease progression in Alzheimer's disease continuum. Given that translocator protein is not exclusively expressed in glia, alternative targets are under rapid development, such as monoamine oxidase B, matrix metalloproteinases, colony-stimulating factor 1 receptor, imidazoline-2 binding sites, cyclooxygenase, cannabinoid-2 receptor, purinergic P2X7 receptor, P2Y12 receptor, the fractalkine receptor, triggering receptor expressed on myeloid cells 2, and receptor for advanced glycation end products. Promising targets should demonstrate a higher specificity for cellular locations with exclusive expression in microglia or astrocyte and activation status (pro- or anti-inflammatory) with highly specific ligand to enable in vivo brain imaging. In this review, we summarised recent advances in the development of neuroinflammation imaging tracers and provided an outlook for promising targets in the future.
Collapse
Affiliation(s)
- Rong Zhou
- Department of Nephrology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bin Ji
- Department of Radiopharmacy and Molecular Imaging, School of Pharmacy, Fudan University, Shanghai, China
| | - Yanyan Kong
- Positron Emission Tomography (PET) Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Limei Qin
- Inner Mongolia Baicaotang Qin Chinese Mongolia Hospital, Hohhot, China
| | - Wuwei Ren
- School of Information Science and Technology, Shanghaitech University, Shanghai, China
| | - Yihui Guan
- Positron Emission Tomography (PET) Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, University of Zurich & Eidgenössische Technische Hochschule Zürich (ETH Zurich), Zurich, Switzerland
| |
Collapse
|