1
|
Williams GK, Akkermans J, Lawson M, Syta P, Staelens S, Adhikari MH, Morton AJ, Nitzsche B, Boltze J, Christou C, Bertoglio D, Ahamed M. Imaging Glucose Metabolism and Dopaminergic Dysfunction in Sheep ( Ovis aries) Brain Using Positron Emission Tomography Imaging Reveals Abnormalities in OVT73 Huntington's Disease Sheep. ACS Chem Neurosci 2024; 15:4082-4091. [PMID: 39420554 DOI: 10.1021/acschemneuro.4c00561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disease that causes cognitive, movement, behavioral, and sleep disturbances, which over time result in progressive disability and eventually death. Clinical translation of novel therapeutics and imaging probes could be accelerated by additional testing in well-characterized large animal models of HD. The major goal of our preliminary cross-sectional study is to demonstrate the feasibility and utility of the unique transgenic sheep model of HD (OVT73) in positron emission tomography (PET) imaging. PET imaging studies were performed in healthy merino sheep (6 year old, n = 3) and OVT73 HD sheep (5.5 year old, n = 3, and 11 year old, n = 3). Region-of-interest and brain atlas labels were defined for regional analyses by using a sheep brain template. [18F]fluorodeoxyglucose ([18F]FDG) was employed to compare the regional brain glucose metabolism and variations in FDG uptake between control and HD sheep. We also used [18F]fluoro-3,4-dihydroxyphenylalanine ([18F]FDOPA) to compare the extent of striatal dysfunction and evaluated the binding potential (BPND) in key brain regions between the groups. Compared with healthy controls and 11 year old HD sheep, the 5.5 year old HD sheep exhibited significantly increased [18F]FDG uptake in several cortical and subcortical brain regions (P < 0.05-0.01). No difference in [18F]FDG uptake was observed between healthy controls and 11 year old HD sheep. Analysis of the [18F]FDOPA BPND parametric maps revealed clusters of reduced binding potential in the 5.5 year old and 11 year old HD sheep compared to the 6 year old control sheep. In this first-of-its-kind study, we showed the usefulness and validity of HD sheep model in imaging cerebral glucose metabolism and dopamine uptake using PET imaging. The identification of discrete patterns of metabolic abnormality using [18F]FDG and decline of [18F]FDOPA uptake may provide a useful means of quantifying early HD-related changes in these models, particularly in the transition from presymptomatic to early symptomatic phases of HD.
Collapse
Affiliation(s)
- Georgia K Williams
- Preclinical, Imaging, and Research Laboratories (PIRL), South Australian Health and Medical Research Institute (SAHMRI), Gilles Plains, Adelaide 5086, Australia
- National Imaging Facility, SAHMRI, Adelaide 5000, Australia
| | - Jordy Akkermans
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp 2160, Belgium
- μNeuro Center for Excellence, University of Antwerp, Antwerp 2160, Belgium
| | - Matt Lawson
- Molecular Imaging and Therapy Research Unit, South Australian Health and Medical Research Institute (SAHMRI), Adelaide 5000, South Australia
| | - Patryk Syta
- Molecular Imaging and Therapy Research Unit, South Australian Health and Medical Research Institute (SAHMRI), Adelaide 5000, South Australia
| | - Steven Staelens
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp 2160, Belgium
- μNeuro Center for Excellence, University of Antwerp, Antwerp 2160, Belgium
| | - Mohit H Adhikari
- μNeuro Center for Excellence, University of Antwerp, Antwerp 2160, Belgium
- Bio-Imaging Lab, University of Antwerp, Antwerp 2160, Belgium
| | - A Jennifer Morton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, U.K
| | - Björn Nitzsche
- Department of Nuclear Medicine, University Hospital Leipzig, Stephanstr. 11, Leipzig 04103, Germany
- Faculty of Veterinary Medicine, Institute of Anatomy, Histology and Embryology, University of Leipzig, An den Tierkliniken 43, Leipzig 04103, Germany
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, U.K
| | - Chris Christou
- Preclinical, Imaging, and Research Laboratories (PIRL), South Australian Health and Medical Research Institute (SAHMRI), Gilles Plains, Adelaide 5086, Australia
- National Imaging Facility, SAHMRI, Adelaide 5000, Australia
| | - Daniele Bertoglio
- μNeuro Center for Excellence, University of Antwerp, Antwerp 2160, Belgium
- Bio-Imaging Lab, University of Antwerp, Antwerp 2160, Belgium
| | - Muneer Ahamed
- Preclinical, Imaging, and Research Laboratories (PIRL), South Australian Health and Medical Research Institute (SAHMRI), Gilles Plains, Adelaide 5086, Australia
- National Imaging Facility, SAHMRI, Adelaide 5000, Australia
- Molecular Imaging and Therapy Research Unit, South Australian Health and Medical Research Institute (SAHMRI), Adelaide 5000, South Australia
- Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, Sydney, New South Wales 2050, Australia
| |
Collapse
|
2
|
Abedi A, Foroutan T, Mohaghegh Shalmani L, Dargahi L. Sex-dependent susceptibility to brain metabolic dysfunction and memory impairment in response to pre and postnatal high-fat diet. J Nutr Biochem 2024; 132:109675. [PMID: 38945454 DOI: 10.1016/j.jnutbio.2024.109675] [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: 02/15/2024] [Revised: 05/15/2024] [Accepted: 05/31/2024] [Indexed: 07/02/2024]
Abstract
The developing brain is sensitive to the impacts of early-life nutritional intake. This study investigates whether maternal high fat diet (HFD) causes glucose metabolism impairment, neuroinflammation, and memory impairment in immature and adult offspring, and whether it may be affected by postweaning diets in a sex-dependent manner in adult offspring. After weaning, female rats were fed HFD (55.9% fat) or normal chow diet (NCD; 10% fat) for 8 weeks before mating, during pregnancy, and lactation. On postnatal day 21 (PND21), the male and female offspring of both groups were split into two new groups, and NCD or HFD feeding was maintained until PND180. On PND21 and PND180, brain glucose metabolism, inflammation, and Alzheimer's pathology-related markers were by qPCR. In adult offspring, peripheral insulin resistance parameters, spatial memory performance, and brain glucose metabolism (18F-FDG-PET scan and protein levels of IDE and GLUT3) were assessed. Histological analysis was also performed on PND21 and adult offspring. On PND21, we found that maternal HFD affected transcript levels of glucose metabolism markers in both sexes. In adult offspring, more profoundly in males, postweaning HFD in combination with maternal HFD induced peripheral and brain metabolic disturbances, impaired memory performance and elevated inflammation, dementia risk markers, and neuronal loss. Our results suggest that maternal HFD affects brain glucose metabolism in the early ages of both sexes. Postweaning HFD sex-dependently causes brain metabolic dysfunction and memory impairment in later-life offspring; effects that can be worsened in combination with maternal HFD.
Collapse
Affiliation(s)
- Azam Abedi
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Tahereh Foroutan
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Leila Mohaghegh Shalmani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Sandhu YK, Bath HS, Shergill J, Liang C, Syed AU, Ngo A, Karim F, Serrano GE, Beach TG, Mukherjee J. [ 18F]Flotaza for Aβ Plaque Diagnostic Imaging: Evaluation in Postmortem Human Alzheimer's Disease Brain Hippocampus and PET/CT Imaging in 5xFAD Transgenic Mice. Int J Mol Sci 2024; 25:7890. [PMID: 39063132 PMCID: PMC11277463 DOI: 10.3390/ijms25147890] [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: 06/16/2024] [Revised: 07/10/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
The diagnostic value of imaging Aβ plaques in Alzheimer's disease (AD) has accelerated the development of fluorine-18 labeled radiotracers with a longer half-life for easier translation to clinical use. We have developed [18F]flotaza, which shows high binding to Aβ plaques in postmortem human AD brain slices with low white matter binding. We report the binding of [18F]flotaza in postmortem AD hippocampus compared to cognitively normal (CN) brains and the evaluation of [18F]flotaza in transgenic 5xFAD mice expressing Aβ plaques. [18F]Flotaza binding was assessed in well-characterized human postmortem brain tissue sections consisting of HP CA1-subiculum (HP CA1-SUB) regions in AD (n = 28; 13 male and 15 female) and CN subjects (n = 32; 16 male and 16 female). Adjacent slices were immunostained with anti-Aβ and analyzed using QuPath. In vitro and in vivo [18F]flotaza PET/CT studies were carried out in 5xFAD mice. Post-mortem human brain slices from all AD subjects were positively IHC stained with anti-Aβ. High [18F]flotaza binding was measured in the HP CA1-SUB grey matter (GM) regions compared to white matter (WM) of AD subjects with GM/WM > 100 in some subjects. The majority of CN subjects had no decipherable binding. Male AD exhibited greater WM than AD females (AD WM♂/WM♀ > 5; p < 0.001) but no difference amongst CN WM. In vitro studies in 5xFAD mice brain slices exhibited high binding [18F]flotaza ratios (>50 versus cerebellum) in the cortex, HP, and thalamus. In vivo, PET [18F]flotaza exhibited binding to Aβ plaques in 5xFAD mice with SUVR~1.4. [18F]Flotaza is a new Aβ plaque PET imaging agent that exhibited high binding to Aβ plaques in postmortem human AD. Along with the promising results in 5xFAD mice, the translation of [18F]flotaza to human PET studies may be worthwhile.
Collapse
Affiliation(s)
- Yasmin K. Sandhu
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA; (Y.K.S.); (H.S.B.); (J.S.); (C.L.); (A.U.S.); (A.N.); (F.K.)
| | - Harman S. Bath
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA; (Y.K.S.); (H.S.B.); (J.S.); (C.L.); (A.U.S.); (A.N.); (F.K.)
| | - Jasmine Shergill
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA; (Y.K.S.); (H.S.B.); (J.S.); (C.L.); (A.U.S.); (A.N.); (F.K.)
| | - Christopher Liang
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA; (Y.K.S.); (H.S.B.); (J.S.); (C.L.); (A.U.S.); (A.N.); (F.K.)
| | - Amina U. Syed
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA; (Y.K.S.); (H.S.B.); (J.S.); (C.L.); (A.U.S.); (A.N.); (F.K.)
| | - Allyson Ngo
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA; (Y.K.S.); (H.S.B.); (J.S.); (C.L.); (A.U.S.); (A.N.); (F.K.)
| | - Fariha Karim
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA; (Y.K.S.); (H.S.B.); (J.S.); (C.L.); (A.U.S.); (A.N.); (F.K.)
| | - Geidy E. Serrano
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (G.E.S.); (T.G.B.)
| | - Thomas G. Beach
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (G.E.S.); (T.G.B.)
| | - Jogeshwar Mukherjee
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA; (Y.K.S.); (H.S.B.); (J.S.); (C.L.); (A.U.S.); (A.N.); (F.K.)
| |
Collapse
|
4
|
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
|
5
|
Salcedo C, Pozo Garcia V, García-Adán B, Ameen AO, Gegelashvili G, Waagepetersen HS, Freude KK, Aldana BI. Increased glucose metabolism and impaired glutamate transport in human astrocytes are potential early triggers of abnormal extracellular glutamate accumulation in hiPSC-derived models of Alzheimer's disease. J Neurochem 2024; 168:822-840. [PMID: 38063257 DOI: 10.1111/jnc.16014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 05/19/2024]
Abstract
Glutamate recycling between neurons and astrocytes is essential to maintain neurotransmitter homeostasis. Disturbances in glutamate homeostasis, resulting in excitotoxicity and neuronal death, have been described as a potential mechanism in Alzheimer's disease (AD) pathophysiology. However, glutamate neurotransmitter metabolism in different human brain cells, particularly astrocytes, has been poorly investigated at the early stages of AD. We sought to investigate glucose and glutamate metabolism in AD by employing human induced pluripotent stem cell (hiPSC)-derived astrocytes and neurons carrying mutations in the amyloid precursor protein (APP) or presenilin-1 (PSEN-1) gene as found in familial types of AD (fAD). Methods such as live-cell bioenergetics and metabolic mapping using [13C]-enriched substrates were used to examine metabolism in the early stages of AD. Our results revealed greater glycolysis and glucose oxidative metabolism in astrocytes and neurons with APP or PSEN-1 mutations, accompanied by an elevated glutamate synthesis compared to control WT cells. Astrocytes with APP or PSEN-1 mutations exhibited reduced expression of the excitatory amino acid transporter 2 (EAAT2), and glutamine uptake increased in mutated neurons, with enhanced glutamate release specifically in neurons with a PSEN-1 mutation. These results demonstrate a hypermetabolic phenotype in astrocytes with fAD mutations possibly linked to toxic glutamate accumulation. Our findings further identify metabolic imbalances that may occur in the early phases of AD pathophysiology.
Collapse
Affiliation(s)
- Claudia Salcedo
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Victoria Pozo Garcia
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bernat García-Adán
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Aishat O Ameen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Georgi Gegelashvili
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Institute of Chemical Biology, Ilia State University, Tbilisi, Georgia
| | - Helle S Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristine K Freude
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Blanca I Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Stanciu GD, Ababei DC, Solcan C, Uritu CM, Craciun VC, Pricope CV, Szilagyi A, Tamba BI. Exploring Cannabinoids with Enhanced Binding Affinity for Targeting the Expanded Endocannabinoid System: A Promising Therapeutic Strategy for Alzheimer's Disease Treatment. Pharmaceuticals (Basel) 2024; 17:530. [PMID: 38675490 PMCID: PMC11053678 DOI: 10.3390/ph17040530] [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: 03/20/2024] [Revised: 04/09/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Despite decades of rigorous research and numerous clinical trials, Alzheimer's disease (AD) stands as a notable healthcare challenge of this century, with effective therapeutic solutions remaining elusive. Recently, the endocannabinoid system (ECS) has emerged as an essential therapeutic target due to its regulatory role in different physiological processes, such as neuroprotection, modulation of inflammation, and synaptic plasticity. This aligns with previous research showing that cannabinoid receptor ligands have the potential to trigger the functional structure of neuronal and brain networks, potentially impacting memory processing. Therefore, our study aims to assess the effects of prolonged, intermittent exposure (over 90 days) to JWH-133 (0.2 mg/kg) and an EU-GMP certified Cannabis sativa L. (Cannabixir® Medium Flos, 2.5 mg/kg) on recognition memory, as well as their influence on brain metabolism and modulation of the expanded endocannabinoid system in APP/PS1 mice. Chronic therapy with cannabinoid receptor ligands resulted in reduced anxiety-like behavior and partially reversed the cognitive deficits. Additionally, a reduction was observed in both the number and size of Aβ plaque deposits, along with decreased cerebral glucose metabolism, as well as a decline in the expression of mTOR and CB2 receptors. Furthermore, the study revealed enlarged astrocytes and enhanced expression of M1 mAChR in mice subjected to cannabinoid treatment. Our findings highlight the pivotal involvement of the extended endocannabinoid system in cognitive decline and pathological aspects associated with AD, presenting essential preclinical evidence to support the continued exploration and assessment of cannabinoid receptor ligands for AD treatment.
Collapse
Affiliation(s)
- Gabriela Dumitrita Stanciu
- Advanced Research and Development Center for Experimental Medicine “Prof. Ostin C. Mungiu”—CEMEX, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (G.D.S.); (A.S.)
| | - Daniela-Carmen Ababei
- Advanced Research and Development Center for Experimental Medicine “Prof. Ostin C. Mungiu”—CEMEX, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (G.D.S.); (A.S.)
- Pharmacodynamics and Clinical Pharmacy Department, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| | - Carmen Solcan
- Faculty of Veterinary Medicine, “Ion Ionescu de la Brad” University of Life Sciences, 700490 Iasi, Romania;
| | - Cristina-Mariana Uritu
- Advanced Research and Development Center for Experimental Medicine “Prof. Ostin C. Mungiu”—CEMEX, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (G.D.S.); (A.S.)
| | - Vlad-Constantin Craciun
- Department of Computer Science, “Alexandru Ioan Cuza” University of Iasi, 700506 Iasi, Romania;
| | - Cosmin-Vasilica Pricope
- Advanced Research and Development Center for Experimental Medicine “Prof. Ostin C. Mungiu”—CEMEX, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (G.D.S.); (A.S.)
| | - Andrei Szilagyi
- Advanced Research and Development Center for Experimental Medicine “Prof. Ostin C. Mungiu”—CEMEX, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (G.D.S.); (A.S.)
| | - Bogdan-Ionel Tamba
- Advanced Research and Development Center for Experimental Medicine “Prof. Ostin C. Mungiu”—CEMEX, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (G.D.S.); (A.S.)
- Department of Pharmacology, Clinical Pharmacology and Algesiology, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| |
Collapse
|
7
|
Chumin EJ, Burton CP, Silvola R, Miner EW, Persohn SC, Veronese M, Territo PR. Brain metabolic network covariance and aging in a mouse model of Alzheimer's disease. Alzheimers Dement 2024; 20:1538-1549. [PMID: 38032015 PMCID: PMC10984484 DOI: 10.1002/alz.13538] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD), the leading cause of dementia worldwide, represents a human and financial impact for which few effective drugs exist to treat the disease. Advances in molecular imaging have enabled assessment of cerebral glycolytic metabolism, and network modeling of brain region have linked to alterations in metabolic activity to AD stage. METHODS We performed 18 F-FDG positron emission tomography (PET) imaging in 4-, 6-, and 12-month-old 5XFAD and littermate controls (WT) of both sexes and analyzed region data via brain metabolic covariance analysis. RESULTS The 5XFAD model mice showed age-related changes in glucose uptake relative to WT mice. Analysis of community structure of covariance networks was different across age and sex, with a disruption of metabolic coupling in the 5XFAD model. DISCUSSION The current study replicates clinical AD findings and indicates that metabolic network covariance modeling provides a translational tool to assess disease progression in AD models.
Collapse
Affiliation(s)
- Evgeny J. Chumin
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Psychological and Brain SciencesIndiana UniversityBloomingtonIndianaUSA
- Indiana University Network Science Institute, Indiana UniversityBloomingtonIndianaUSA
| | - Charles P. Burton
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Rebecca Silvola
- Department of MedicineDivision of Clinical PharmacologyIndiana University School of MedicineIndianapolisIndianaUSA
- Eli Lilly and CompanyIndianapolisIndianaUSA
| | - Ethan W. Miner
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Scott C. Persohn
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Mattia Veronese
- Department of Information EngineeringUniversity of PaduaPaduaItaly
- Department of NeuroimagingKing's College LondonLondonUK
| | - Paul R. Territo
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of MedicineDivision of Clinical PharmacologyIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
8
|
Kong Y, Cao L, Xie F, Wang X, Zuo C, Shi K, Rominger A, Huang Q, Xiao J, Jiang D, Guan Y, Ni R. Reduced SV2A and GABA A receptor levels in the brains of type 2 diabetic rats revealed by [ 18F]SDM-8 and [ 18F]flumazenil PET. Biomed Pharmacother 2024; 172:116252. [PMID: 38325265 DOI: 10.1016/j.biopha.2024.116252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/19/2023] [Accepted: 02/02/2024] [Indexed: 02/09/2024] Open
Abstract
PURPOSE Type 2 diabetes mellitus (T2DM) is associated with a greater risk of Alzheimer's disease. Synaptic impairment and protein aggregates have been reported in the brains of T2DM models. Here, we assessed whether neurodegenerative changes in synaptic vesicle 2 A (SV2A), γ-aminobutyric acid type A (GABAA) receptor, amyloid-β, tau and receptor for advanced glycosylation end product (RAGE) can be detected in vivo in T2DM rats. METHODS Positron emission tomography (PET) using [18F]SDM-8 (SV2A), [18F]flumazenil (GABAA receptor), [18F]florbetapir (amyloid-β), [18F]PM-PBB3 (tau), and [18F]FPS-ZM1 (RAGE) was carried out in 12-month-old diabetic Zucker diabetic fatty (ZDF) and SpragueDawley (SD) rats. Immunofluorescence staining, Thioflavin S staining, proteomic profiling and pathway analysis were performed on the brain tissues of ZDF and SD rats. RESULTS Reduced cortical [18F]SDM-8 uptake and cortical and hippocampal [18F]flumazenil uptake were observed in 12-month-old ZDF rats compared to SD rats. The regional uptake of [18F]florbetapir and [18F]PM-PBB3 was comparable in the brains of 12-month-old ZDF and SD rats. Immunofluorescence staining revealed Thioflavin S-negative, phospho-tau-positive inclusions in the cortex and hypothalamus in the brains of ZDF rats and the absence of amyloid-beta deposits. The level of GABAA receptors was lower in the cortex of ZDF rats than SD rats. Proteomic analysis further demonstrated that, compared with SD rats, synaptic-related proteins and pathways were downregulated in the hippocampus of ZDF rats. CONCLUSION These findings provide in vivo evidence for regional reductions in SV2A and GABAA receptor levels in the brains of aged T2DM ZDF rats.
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
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiuzhe Wang
- Dept. Neurology, Shanghai Sixth People's Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chuantao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Kuangyu Shi
- Dept. Nuclear Medicine, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Axel Rominger
- Dept. Nuclear Medicine, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Qi Huang
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianfei Xiao
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Donglang Jiang
- 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, Inselspital, Bern University Hospital, Bern, Switzerland; Inst. Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
9
|
Jullienne A, Szu JI, Quan R, Trinh MV, Norouzi T, Noarbe BP, Bedwell AA, Eldridge K, Persohn SC, Territo PR, Obenaus A. Cortical cerebrovascular and metabolic perturbations in the 5xFAD mouse model of Alzheimer's disease. Front Aging Neurosci 2023; 15:1220036. [PMID: 37533765 PMCID: PMC10392850 DOI: 10.3389/fnagi.2023.1220036] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/03/2023] [Indexed: 08/04/2023] Open
Abstract
Introduction The 5xFAD mouse is a popular model of familial Alzheimer's disease (AD) that is characterized by early beta-amyloid (Aβ) deposition and cognitive decrements. Despite numerous studies, the 5xFAD mouse has not been comprehensively phenotyped for vascular and metabolic perturbations over its lifespan. Methods Male and female 5xFAD and wild type (WT) littermates underwent in vivo 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) imaging at 4, 6, and 12 months of age to assess regional glucose metabolism. A separate cohort of mice (4, 8, 12 months) underwent "vessel painting" which labels all cerebral vessels and were analyzed for vascular characteristics such as vessel density, junction density, vessel length, network complexity, number of collaterals, and vessel diameter. Results With increasing age, vessels on the cortical surface in both 5xFAD and WT mice showed increased vessel length, vessel and junction densities. The number of collateral vessels between the middle cerebral artery (MCA) and the anterior and posterior cerebral arteries decreased with age but collateral diameters were significantly increased only in 5xFAD mice. MCA total vessel length and junction density were decreased in 5xFAD mice compared to WT at 4 months. Analysis of 18F-FDG cortical uptake revealed significant differences between WT and 5xFAD mice spanning 4-12 months. Broadly, 5xFAD males had significantly increased 18F-FDG uptake at 12 months compared to WT mice. In most cortical regions, female 5xFAD mice had reduced 18F-FDG uptake compared to WT across their lifespan. Discussion While the 5xFAD mouse exhibits AD-like cognitive deficits as early as 4 months of age that are associated with increasing Aβ deposition, we only found significant differences in cortical vascular features in males, not in females. Interestingly, 5xFAD male and female mice exhibited opposite effects in 18F-FDG uptake. The MCA supplies blood to large portions of the somatosensory cortex and portions of motor and visual cortex and increased vessel length alongside decreased collaterals which coincided with higher metabolic rates in 5xFAD mice. Thus, a potential mismatch between metabolic demand and vascular delivery of nutrients in the face of increasing Aβ deposition could contribute to the progressive cognitive deficits seen in the 5xFAD mouse model.
Collapse
Affiliation(s)
- Amandine Jullienne
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Jenny I. Szu
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Ryan Quan
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Michelle V. Trinh
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Tannoz Norouzi
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Brenda P. Noarbe
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Amanda A. Bedwell
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Kierra Eldridge
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Scott C. Persohn
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Paul R. Territo
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, Indianapolis, IN, United States
- Department of Medicine, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Andre Obenaus
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
10
|
Wang YY, Zhou YN, Jiang L, Wang S, Zhu L, Zhang SS, Yang H, He Q, Liu L, Xie YH, Liang X, Tang J, Chao FL, Tang Y. Long-term voluntary exercise inhibited AGE/RAGE and microglial activation and reduced the loss of dendritic spines in the hippocampi of APP/PS1 transgenic mice. Exp Neurol 2023; 363:114371. [PMID: 36871860 DOI: 10.1016/j.expneurol.2023.114371] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/22/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
Alzheimer's disease (AD) is closely related to hippocampal synapse loss, which can be alleviated by running exercise. However, further studies are needed to determine whether running exercise reduces synapse loss in the hippocampus in an AD model by regulating microglia. Ten-month-old male wild-type mice and APP/PS1 mice were randomly divided into control and running groups. All mice in the running groups were subjected to voluntary running exercise for four months. After the behavioral tests, immunohistochemistry, stereological methods, immunofluorescence staining, 3D reconstruction, western blotting and RNA-Seq were performed. Running exercise improved the spatial learning and memory abilities of APP/PS1 mice and increased the total number of dendritic spines, the levels of the PSD-95 and Synapsin Ia/b proteins, the colocalization of PSD-95 and neuronal dendrites (MAP-2) and the number of PSD-95-contacting astrocytes (GFAP) in the hippocampi of APP/PS1 mice. Moreover, running exercise reduced the relative expression of CD68 and Iba-1, the number of Iba-1+ microglia and the colocalization of PSD-95 and Iba-1+ microglia in the hippocampi of APP/PS1 mice. The RNA-Seq results showed that some differentially expressed genes (DEGs) related to the complement system (Cd59b, Serping1, Cfh, A2m, and Trem2) were upregulated in the hippocampi of APP/PS1 mice, while running exercise downregulated the C3 gene. At the protein level, running exercise also reduced the expression of advanced glycation end products (AGEs), receptor for advanced glycation end products (RAGE), C1q and C3 in the hippocampus and AGEs and RAGE in hippocampal microglia in APP/PS1 mice. Furthermore, the Col6a3, Scn5a, Cxcl5, Tdg and Clec4n genes were upregulated in the hippocampi of APP/PS1 mice but downregulated after running, and these genes were associated with the C3 and RAGE genes according to protein-protein interaction (PPI) analysis. These findings indicate that long-term voluntary exercise might protect hippocampal synapses and affect the function and activation of microglia, the AGE/RAGE signaling pathway in microglia and the C1q/C3 complement system in the hippocampus in APP/PS1 mice, and these effects may be related to the Col6a3, Scn5a, Cxcl5, Tdg and Clec4n genes. The current results provide an important basis for identifying targets for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Yi-Ying Wang
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Yu-Ning Zhou
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Lin Jiang
- Lab Teaching & Management Center, Chongqing Medical University, Chongqing 400016, PR China
| | - Shun Wang
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Lin Zhu
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Shan-Shan Zhang
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Hao Yang
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Qi He
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Li Liu
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Yu-Han Xie
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Xin Liang
- Department of Pathophysiology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Jing Tang
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Feng-Lei Chao
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China.
| | - Yong Tang
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
11
|
Lansdell TA, Xu H, Galligan JJ, Dorrance AM. Effects of Striatal Amyloidosis on the Dopaminergic System and Behavior: A Comparative Study in Male and Female 5XFAD Mice. J Alzheimers Dis 2023; 94:1361-1375. [PMID: 37424461 DOI: 10.3233/jad-220905] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
BACKGROUND Nearly two-thirds of patients diagnosed with Alzheimer's disease (AD) are female. In addition, female patients with AD have more significant cognitive impairment than males at the same disease stage. This disparity suggests there are sex differences in AD progression. While females appear to be more affected by AD, most published behavioral studies utilize male mice. In humans, there is an association between antecedent attention-deficit/hyperactivity disorder and increased risk of dementia. Functional connectivity studies indicate that dysfunctional cortico-striatal networks contribute to hyperactivity in attention deficit hyperactivity disorder. Higher plaque density in the striatum accurately predicts the presence of clinical AD pathology. In addition, there is a link between AD-related memory dysfunction and dysfunctional dopamine signaling. OBJECTIVE With the need to consider sex as a biological variable, we investigated the influence of sex on striatal plaque burden, dopaminergic signaling, and behavior in prodromal 5XFAD mice. METHODS Six-month-old male and female 5XFAD and C57BL/6J mice were evaluated for striatal amyloid plaque burden, locomotive behavior, and changes in dopaminergic machinery in the striatum. RESULTS 5XFAD female mice had a higher striatal amyloid plaque burden than male 5XFAD mice. 5XFAD females, but not males, were hyperactive. Hyperactivity in female 5XFAD mice was associated with increased striatal plaque burden and changes in dopamine signaling in the dorsal striatum. CONCLUSION Our results indicate that the progression of amyloidosis involves the striatum in females to a greater extent than in males. These studies have significant implications for using male-only cohorts in the study of AD progression.
Collapse
Affiliation(s)
- Theresa A Lansdell
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - Hui Xu
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - James J Galligan
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
12
|
Solas M, Zamarbide M, Ardanaz CG, Ramírez MJ, Pérez-Mediavilla A. The Cognitive Improvement and Alleviation of Brain Hypermetabolism Caused by FFAR3 Ablation in Tg2576 Mice Is Persistent under Diet-Induced Obesity. Int J Mol Sci 2022; 23:13591. [PMID: 36362376 PMCID: PMC9654726 DOI: 10.3390/ijms232113591] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/28/2022] [Accepted: 11/03/2022] [Indexed: 11/10/2022] Open
Abstract
Obesity and aging are becoming increasingly prevalent across the globe. It has been established that aging is the major risk factor for Alzheimer's disease (AD), and it is becoming increasingly evident that obesity and the associated insulin resistance are also notably relevant risk factors. The biological plausibility of the link between high adiposity, insulin resistance, and dementia is central for understanding AD etiology, and to form bases for prevention efforts to decrease the disease burden. Several studies have demonstrated a strong association between short chain fatty acid receptor FFAR3 and insulin sensitivity. Interestingly, it has been recently established that FFAR3 mRNA levels are increased in early stages of the AD pathology, indicating that FFAR3 could play a key role in AD onset and progression. Indeed, in the present study we demonstrate that the ablation of the Ffar3 gene in Tg2576 mice prevents the development of cognitive deficiencies in advanced stages of the disease. Notably, this cognitive improvement is also maintained upon a severe metabolic challenge such as the exposure to high-fat diet (HFD) feeding. Moreover, FFAR3 deletion restores the brain hypermetabolism displayed by Tg2576 mice. Collectively, these data postulate FFAR3 as a potential novel target for AD.
Collapse
Affiliation(s)
- Maite Solas
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Marta Zamarbide
- Neuroscience Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
| | - Carlos G. Ardanaz
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - María J. Ramírez
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Alberto Pérez-Mediavilla
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Neuroscience Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain
| |
Collapse
|
13
|
Venkataraman AV, Mansur A, Rizzo G, Bishop C, Lewis Y, Kocagoncu E, Lingford-Hughes A, Huiban M, Passchier J, Rowe JB, Tsukada H, Brooks DJ, Martarello L, Comley RA, Chen L, Schwarz AJ, Hargreaves R, Gunn RN, Rabiner EA, Matthews PM. Widespread cell stress and mitochondrial dysfunction occur in patients with early Alzheimer's disease. Sci Transl Med 2022; 14:eabk1051. [PMID: 35976998 DOI: 10.1126/scitranslmed.abk1051] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cell stress and impaired oxidative phosphorylation are central to mechanisms of synaptic loss and neurodegeneration in the cellular pathology of Alzheimer's disease (AD). In this study, we quantified the in vivo expression of the endoplasmic reticulum stress marker, sigma 1 receptor (S1R), using [11C]SA4503 positron emission tomography (PET), the mitochondrial complex I (MC1) with [18F]BCPP-EF, and the presynaptic vesicular protein SV2A with [11C]UCB-J in 12 patients with early AD and in 16 cognitively normal controls. We integrated these molecular measures with assessments of regional brain volumes and cerebral blood flow (CBF) measured with magnetic resonance imaging arterial spin labeling. Eight patients with AD were followed longitudinally to estimate the rate of change of the physiological and structural pathology markers with disease progression. The patients showed widespread increases in S1R (≤ 27%) and regional reduction in MC1 (≥ -28%) and SV2A (≥ -25%) radioligand binding, brain volume (≥ -23%), and CBF (≥ -26%). [18F]BCPP-EF PET MC1 binding (≥ -12%) and brain volumes (≥ -5%) showed progressive reductions over 12 to 18 months, suggesting that they both could be used as pharmacodynamic indicators in early-stage therapeutics trials. Associations of reduced MC1 and SV2A and increased S1R radioligand binding with reduced cognitive performance in AD, although exploratory, suggested a loss of metabolic functional reserve with disease. Our study thus provides in vivo evidence for widespread, clinically relevant cellular stress and bioenergetic abnormalities in early AD.
Collapse
Affiliation(s)
- Ashwin V Venkataraman
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK.,UK Dementia Research Institute at Imperial College London, London W12 0NN, UK
| | | | - Gaia Rizzo
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK.,Invicro LLC, London W12 0NN, UK
| | | | | | | | | | | | | | | | - Hideo Tsukada
- Hamamatsu Photonics, Hamakita, Hamamatsu, Shizuoka 4348601, Japan
| | - David J Brooks
- University of Newcastle upon Tyne, Newcastle NE2 4HH, UK.,Department of Clinical Medicine, Aarhus University, Aarhus 8200, Denmark
| | | | | | | | | | | | - Roger N Gunn
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK.,Invicro LLC, London W12 0NN, UK
| | - Eugenii A Rabiner
- Invicro LLC, London W12 0NN, UK.,King's College London, London SE5 8AF, UK
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK.,UK Dementia Research Institute at Imperial College London, London W12 0NN, UK
| |
Collapse
|
14
|
Oh SJ, Lee N, Nam KR, Kang KJ, Han SJ, Lee KC, Lee YJ, Choi JY. Amyloid pathology induces dysfunction of systemic neurotransmission in aged APPswe/PS2 mice. Front Neurosci 2022; 16:930613. [PMID: 35992913 PMCID: PMC9389227 DOI: 10.3389/fnins.2022.930613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/15/2022] [Indexed: 11/21/2022] Open
Abstract
This study aimed to investigate how amyloid pathology affects the functional aspects of neurotransmitter systems in Alzheimer’s disease. APPswe/PS2 mice (21 months of age) and wild-type (WT) mice underwent positron emission tomography (PET) and magnetic resonance spectroscopy (MRS). First, we obtained 18F-FDG and 18F-florbetaben PET scans to evaluate neuronal integrity and amyloid pathology. Second, 18F-FPEB and 18F-FMZ PET data were acquired to assess the excitatory-inhibitory neurotransmission. Third, to monitor the dopamine system, 18F-fallypride PET was performed. Amyloid PET imaging revealed that radioactivity was higher in the AD group than that in the WT group, which was validated by immunohistochemistry. In the cortical and limbic areas, the AD group showed a 25–27% decrease and 14–35% increase in the glutamatergic and GABAergic systems, respectively. The dopaminergic system in the AD group exhibited a 29% decrease in brain uptake compared with that in the WT group. A reduction in glutamate, N-acetylaspartate, and taurine levels was observed in the AD group using MRS. Our results suggest that dysfunction of the neurotransmitter system is associated with AD pathology. Among the systems, the GABAergic system was prominent, implying that the inhibitory neurotransmission system may be the most vulnerable to AD pathology.
Collapse
Affiliation(s)
- Se Jong Oh
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Namhun Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Kyung Rok Nam
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Kyung Jun Kang
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Sang Jin Han
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Kyo Chul Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Yong Jin Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Jae Yong Choi
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
- Radiological and Medico-Oncological Sciences, University of Science and Technology (UST), Seoul, South Korea
- *Correspondence: Jae Yong Choi,
| |
Collapse
|
15
|
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: 1.0] [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
|
16
|
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: 0] [Impact Index Per Article: 0] [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
|
17
|
Zhang SS, Zhu L, Peng Y, Zhang L, Chao FL, Jiang L, Xiao Q, Liang X, Tang J, Yang H, He Q, Guo YJ, Zhou CN, Tang Y. Long-term running exercise improves cognitive function and promotes microglial glucose metabolism and morphological plasticity in the hippocampus of APP/PS1 mice. J Neuroinflammation 2022; 19:34. [PMID: 35123512 PMCID: PMC8817568 DOI: 10.1186/s12974-022-02401-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 01/24/2022] [Indexed: 02/06/2023] Open
Abstract
Background The role of physical exercise in the prevention of Alzheimer’s disease (AD) has been widely studied. Microglia play an important role in AD. Triggering receptor expressed in myeloid cells 2 (TREM2) is expressed on microglia and is known to mediate microglial metabolic activity and brain glucose metabolism. However, the relationship between brain glucose metabolism and microglial metabolic activity during running exercise in APP/PS1 mice remains unclear. Methods Ten-month-old male APP/PS1 mice and wild-type mice were randomly divided into sedentary groups or running groups (AD_Sed, WT_Sed, AD_Run and WT_Run, n = 20/group). Running mice had free access to a running wheel for 3 months. Behavioral tests, [18]F-FDG-PET and hippocampal RNA-Seq were performed. The expression levels of microglial glucose transporter (GLUT5), TREM2, soluble TREM2 (sTREM2), TYRO protein tyrosine kinase binding protein (TYROBP), secreted phosphoprotein 1 (SPP1), and phosphorylated spleen tyrosine kinase (p-SYK) were estimated by western blot or ELISA. Immunohistochemistry, stereological methods and immunofluorescence were used to investigate the morphology, proliferation and activity of microglia. Results Long-term voluntary running significantly improved cognitive function in APP/PS1 mice. Although there were few differentially expressed genes (DEGs), gene set enrichment analysis (GSEA) showed enriched glycometabolic pathways in APP/PS1 running mice. Running exercise increased FDG uptake in the hippocampus of APP/PS1 mice, as well as the protein expression of GLUT5, TREM2, SPP1 and p-SYK. The level of sTREM2 decreased in the plasma of APP/PS1 running mice. The number of microglia, the length and endpoints of microglial processes, and the ratio of GLUT5+/IBA1+ microglia were increased in the dentate gyrus (DG) of APP/PS1 running mice. Running exercise did not alter the number of 5-bromo-2′-deoxyuridine (BrdU)+/IBA1+ microglia but reduced the immunoactivity of CD68 in the hippocampus of APP/PS1 mice. Conclusions Running exercise inhibited TREM2 shedding and maintained TREM2 protein levels, which were accompanied by the promotion of brain glucose metabolism, microglial glucose metabolism and morphological plasticity in the hippocampus of AD mice. Microglia might be a structural target responsible for the benefits of running exercise in AD. Promoting microglial glucose metabolism and morphological plasticity modulated by TREM2 might be a novel strategy for AD treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02401-5.
Collapse
|
18
|
Neuroimaging of Mouse Models of Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10020305. [PMID: 35203515 PMCID: PMC8869427 DOI: 10.3390/biomedicines10020305] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 12/23/2022] Open
Abstract
Magnetic resonance imaging (MRI) and positron emission tomography (PET) have made great strides in the diagnosis and our understanding of Alzheimer’s Disease (AD). Despite the knowledge gained from human studies, mouse models have and continue to play an important role in deciphering the cellular and molecular evolution of AD. MRI and PET are now being increasingly used to investigate neuroimaging features in mouse models and provide the basis for rapid translation to the clinical setting. Here, we provide an overview of the human MRI and PET imaging landscape as a prelude to an in-depth review of preclinical imaging in mice. A broad range of mouse models recapitulate certain aspects of the human AD, but no single model simulates the human disease spectrum. We focused on the two of the most popular mouse models, the 3xTg-AD and the 5xFAD models, and we summarized all known published MRI and PET imaging data, including contrasting findings. The goal of this review is to provide the reader with broad framework to guide future studies in existing and future mouse models of AD. We also highlight aspects of MRI and PET imaging that could be improved to increase rigor and reproducibility in future imaging studies.
Collapse
|
19
|
Kim D, Jo YS, Jo HS, Bae S, Kwon YW, Oh YS, Yoon JH. Comparative Phosphoproteomics of Neuro-2a Cells under Insulin Resistance Reveals New Molecular Signatures of Alzheimer's Disease. Int J Mol Sci 2022; 23:ijms23021006. [PMID: 35055191 PMCID: PMC8781554 DOI: 10.3390/ijms23021006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/03/2022] [Accepted: 01/12/2022] [Indexed: 12/29/2022] Open
Abstract
Insulin in the brain is a well-known critical factor in neuro-development and regulation of adult neurogenesis in the hippocampus. The abnormality of brain insulin signaling is associated with the aging process and altered brain plasticity, and could promote neurodegeneration in the late stage of Alzheimer’s disease (AD). The precise molecular mechanism of the relationship between insulin resistance and AD remains unclear. The development of phosphoproteomics has advanced our knowledge of phosphorylation-mediated signaling networks and could elucidate the molecular mechanisms of certain pathological conditions. Here, we applied a reliable phosphoproteomic approach to Neuro2a (N2a) cells to identify their molecular features under two different insulin-resistant conditions with clinical relevance: inflammation and dyslipidemia. Despite significant difference in overall phosphoproteome profiles, we found molecular signatures and biological pathways in common between two insulin-resistant conditions. These include the integrin and adenosine monophosphate-activated protein kinase pathways, and we further verified these molecular targets by subsequent biochemical analysis. Among them, the phosphorylation levels of acetyl-CoA carboxylase and Src were reduced in the brain from rodent AD model 5xFAD mice. This study provides new molecular signatures for insulin resistance in N2a cells and possible links between the molecular features of insulin resistance and AD.
Collapse
Affiliation(s)
- Dayea Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), Daegu 41061, Korea;
| | - Yeon Suk Jo
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Korea; (Y.S.J.); (H.-S.J.); (S.B.); (Y.W.K.)
- Department of Brain-Cognitive Science, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Han-Seul Jo
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Korea; (Y.S.J.); (H.-S.J.); (S.B.); (Y.W.K.)
| | - Sungwon Bae
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Korea; (Y.S.J.); (H.-S.J.); (S.B.); (Y.W.K.)
| | - Yang Woo Kwon
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Korea; (Y.S.J.); (H.-S.J.); (S.B.); (Y.W.K.)
| | - Yong-Seok Oh
- Department of Brain-Cognitive Science, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
- Correspondence: (Y.-S.O.); (J.H.Y.); Tel.: +82-53-785-6114 (Y.-S.O.); +82-53-980-8341 (J.H.Y.)
| | - Jong Hyuk Yoon
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Korea; (Y.S.J.); (H.-S.J.); (S.B.); (Y.W.K.)
- Correspondence: (Y.-S.O.); (J.H.Y.); Tel.: +82-53-785-6114 (Y.-S.O.); +82-53-980-8341 (J.H.Y.)
| |
Collapse
|
20
|
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: 3] [Impact Index Per Article: 1.5] [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
|
21
|
Bouter C, Irwin C, Franke TN, Beindorff N, Bouter Y. Quantitative Brain Positron Emission Tomography in Female 5XFAD Alzheimer Mice: Pathological Features and Sex-Specific Alterations. Front Med (Lausanne) 2021; 8:745064. [PMID: 34901060 PMCID: PMC8661108 DOI: 10.3389/fmed.2021.745064] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/04/2021] [Indexed: 11/13/2022] Open
Abstract
Successful back-translating clinical biomarkers and molecular imaging methods of Alzheimer's disease (AD), including positron emission tomography (PET), are very valuable for the evaluation of new therapeutic strategies and increase the quality of preclinical studies. 18F-Fluorodeoxyglucose (FDG)–PET and 18F-Florbetaben–PET are clinically established biomarkers capturing two key pathological features of AD. However, the suitability of 18F-FDG– and amyloid–PET in the widely used 5XFAD mouse model of AD is still unclear. Furthermore, only data on male 5XFAD mice have been published so far, whereas studies in female mice and possible sex differences in 18F-FDG and 18F-Florbetaben uptake are missing. The aim of this study was to evaluate the suitability of 18F-FDG– and 18F-Florbetaben–PET in 7-month-old female 5XFAD and to assess possible sex differences between male and female 5XFAD mice. We could demonstrate that female 5XFAD mice showed a significant reduction in brain glucose metabolism and increased cerebral amyloid deposition compared with wild type animals, in accordance with the pathology seen in AD patients. Furthermore, we showed for the first time that the hypometabolism in 5XFAD mice is gender-dependent and more pronounced in female mice. Therefore, these results support the feasibility of small animal PET imaging with 18F-FDG- and 18F-Florbetaben in 5XFAD mice in both, male and female animals. Moreover, our findings highlight the need to account for sex differences in studies working with 5XFAD mice.
Collapse
Affiliation(s)
- Caroline Bouter
- Department of Nuclear Medicine, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Caroline Irwin
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Timon N Franke
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Nicola Beindorff
- Berlin Experimental Radionuclide Imaging Center (BERIC), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Yvonne Bouter
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| |
Collapse
|
22
|
Demetrius LA, Eckert A, Grimm A. Sex differences in Alzheimer's disease: metabolic reprogramming and therapeutic intervention. Trends Endocrinol Metab 2021; 32:963-979. [PMID: 34654630 DOI: 10.1016/j.tem.2021.09.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/05/2021] [Accepted: 09/21/2021] [Indexed: 10/20/2022]
Abstract
Studies on the sporadic form of Alzheimer's disease (AD) have revealed three classes of risk factor: age, genetics, and sex. These risk factors point to a metabolic dysregulation as the origin of AD. Adaptive alterations in cerebral metabolism are the rationale for the Metabolic Reprogramming (MR) Theory of the origin of AD. The theory contends that the progression toward AD involves three adaptive events: a hypermetabolic phase, a prolonged prodromal phase, and a metabolic collapse. This article exploits the MR Theory to elucidate the effect of hormonal changes on the origin and progression of AD in women. The theory invokes bioenergetic signatures of the menopausal transition to propose sex-specific diagnostic program and therapeutic strategies.
Collapse
Affiliation(s)
- Lloyd A Demetrius
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Anne Eckert
- University of Basel, Transfaculty Research Platform Molecular and Cognitive Neuroscience, 4002 Basel, Switzerland; Neurobiology Lab for Brain Aging and Mental Health, Psychiatric University Clinics, 4002 Basel, Switzerland
| | - Amandine Grimm
- University of Basel, Transfaculty Research Platform Molecular and Cognitive Neuroscience, 4002 Basel, Switzerland; Neurobiology Lab for Brain Aging and Mental Health, Psychiatric University Clinics, 4002 Basel, Switzerland; University of Basel, Life Sciences Training Facility, 4055 Basel, Switzerland.
| |
Collapse
|
23
|
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: 3.3] [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
|
24
|
Tataryn NM, Singh V, Dyke JP, Berk-Rauch HE, Clausen DM, Aronowitz E, Norris EH, Strickland S, Ahn HJ. Vascular endothelial growth factor associated dissimilar cerebrovascular phenotypes in two different mouse models of Alzheimer's Disease. Neurobiol Aging 2021; 107:96-108. [PMID: 34416494 PMCID: PMC8595520 DOI: 10.1016/j.neurobiolaging.2021.07.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 01/14/2023]
Abstract
Vascular perturbations and cerebral hypometabolism are emerging as important components of Alzheimer's disease (AD). While various in vivo imaging modalities have been designed to detect changes of cerebral perfusion and metabolism in AD patients and animal models, study results were often heterogenous with respect to imaging techniques and animal models. We therefore evaluated cerebral perfusion and glucose metabolism of two popular transgenic AD mouse strains, TgCRND8 and 5xFAD, at 7 and 12 months-of-age under identical conditions and analyzed possible molecular mechanisms underlying heterogeneous cerebrovascular phenotypes. Results revealed disparate findings in these two strains, displaying important aspects of AD progression. TgCRND8 mice showed significantly decreased cerebral blood flow and glucose metabolism with unchanged cerebral blood volume (CBV) at 12 months-of-age whereas 5xFAD mice showed unaltered glucose metabolism with significant increase in CBV at 12 months-of-age and a biphasic pattern of early hypoperfusion followed by a rebound to normal cerebral blood flow in late disease. Finally, immunoblotting assays suggested that VEGF dependent vascular tone change may restore normoperfusion and increase CBV in 5xFAD.
Collapse
Affiliation(s)
- Nicholas M Tataryn
- Tri-Institutional Training Program in Laboratory Animal Medicine and Science, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, and The Rockefeller University, New York, New York, USA and Center for Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, Rockefeller University, New York, NY, USA; Division of Comparative Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Vishal Singh
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Jonathan P Dyke
- Citigroup Biomedical Imaging Center, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Hanna E Berk-Rauch
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, Rockefeller University, New York, NY, USA
| | - Dana M Clausen
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Eric Aronowitz
- Citigroup Biomedical Imaging Center, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Erin H Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, Rockefeller University, New York, NY, USA
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, Rockefeller University, New York, NY, USA
| | - Hyung Jin Ahn
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, USA; Brain Health Institute, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
25
|
Huang J, Lai JHC, Tse KH, Cheng GWY, Liu Y, Chen Z, Han X, Chen L, Xu J, Chan KWY. Deep neural network based CEST and AREX processing: Application in imaging a model of Alzheimer's disease at 3 T. Magn Reson Med 2021; 87:1529-1545. [PMID: 34657318 DOI: 10.1002/mrm.29044] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/26/2021] [Accepted: 09/22/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE To optimize and apply deep neural network based CEST (deepCEST) and apparent exchange dependent-relaxation (deepAREX) for imaging the mouse brain with Alzheimer's disease (AD) at 3T MRI. METHODS CEST and T1 data of central and anterior brain slices of 10 AD mice and 10 age-matched wild type (WT) mice were acquired at a 3T animal MRI scanner. The networks of deepCEST/deepAREX were optimized and trained on the WT data. The CEST/AREX contrasts of AD and WT mice predicted by the networks were analyzed and further validated by immunohistochemistry. RESULTS After optimization and training on CEST data of WT mice, deepCEST/deepAREX could rapidly (~1 s) generate precise CEST and AREX results for unseen CEST data of AD mice, indicating the accuracy and generalization of the networks. Significant lower amide weighted (3.5 ppm) signal related to amyloid β-peptide (Aβ) plaque depositions, which was validated by immunohistochemistry results, was detected in both central and anterior brain slices of AD mice compared to WT mice. Decreased magnetization transfer (MT) signal was also found in AD mice especially in the anterior slice. CONCLUSION DeepCEST/deepAREX could rapidly generate accurate CEST/AREX contrasts in animal study. The well-optimized deepCEST/deepAREX have potential for AD differentiation at 3T MRI.
Collapse
Affiliation(s)
- Jianpan Huang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Joseph H C Lai
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Kai-Hei Tse
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Gerald W Y Cheng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Yang Liu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China.,Hong Kong Centre for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, China
| | - Zilin Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Xiongqi Han
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Lin Chen
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, Maryland, USA.,Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, China
| | - Jiadi Xu
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, Maryland, USA.,Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kannie W Y Chan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China.,Hong Kong Centre for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, China.,Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
26
|
Choi H, Choi Y, Lee EJ, Kim H, Lee Y, Kwon S, Hwang DW, Lee DS. Hippocampal glucose uptake as a surrogate of metabolic change of microglia in Alzheimer's disease. J Neuroinflammation 2021; 18:190. [PMID: 34465358 PMCID: PMC8408933 DOI: 10.1186/s12974-021-02244-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/18/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Dynamically altered microglia play an important role in the progression of Alzheimer's disease (AD). Here, we found a close association of the metabolic reconfiguration of microglia with increased hippocampal glucose uptake on [18F]fluorodeoxyglucose (FDG) PET. METHODS We used an AD animal model, 5xFAD, to analyze hippocampal glucose metabolism using both animal FDG PET and ex vivo FDG uptake test. Cells of the hippocampus were isolated to perform single-cell RNA-sequencing (scRNA-seq). The molecular features of cells associated with glucose metabolism were analyzed at a single-cell level. In order to apply our findings to human brain imaging study, brain FDG PET data obtained from the Alzheimer's Disease Neuroimaging Initiative were analyzed. FDG uptake in the hippocampus was compared according to the diagnosis, AD, mild cognitive impairment, and controls. The correlation analysis between hippocampal FDG uptake and soluble TREM2 in cerebrospinal fluid was performed. RESULTS In the animal study, 8- and 12-month-old 5xFAD mice showed higher FDG uptake in the hippocampus than wild-type mice. Cellular FDG uptake tests showed that FDG activity in hippocampal microglia was increased in the AD model, while FDG activity in non-microglial cells of the hippocampus was not different between the AD model and wild-type. scRNA-seq data showed that changes in glucose metabolism signatures including glucose transporters, glycolysis and oxidative phosphorylation, mainly occurred in microglia. A subset of microglia with higher glucose transporters with defective glycolysis and oxidative phosphorylation was increased according to disease progression. In the human imaging study, we found a positive association between soluble TREM2 and hippocampal FDG uptake. FDG uptake in the hippocampus at the baseline scan predicted mild cognitive impairment conversion to AD. CONCLUSIONS We identified the reconfiguration of microglial glucose metabolism in the hippocampus of AD, which could be evaluated by FDG PET as a feasible surrogate imaging biomarker for microglia-mediated inflammation.
Collapse
Affiliation(s)
- Hongyoon Choi
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Seoul, Jongo-Gu 03080 Republic of Korea
| | - Yoori Choi
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
| | - Eun Ji Lee
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
| | - Hyun Kim
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
| | - Youngsun Lee
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
| | - Seokjun Kwon
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
| | - Do Won Hwang
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
| | - Dong Soo Lee
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Seoul, Jongo-Gu 03080 Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
| | - for the Alzheimer’s Disease Neuroimaging Initiative
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Seoul, Jongo-Gu 03080 Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
| |
Collapse
|
27
|
Franke TN, Irwin C, Bayer TA, Brenner W, Beindorff N, Bouter C, Bouter Y. In vivo Imaging With 18F-FDG- and 18F-Florbetaben-PET/MRI Detects Pathological Changes in the Brain of the Commonly Used 5XFAD Mouse Model of Alzheimer's Disease. Front Med (Lausanne) 2020; 7:529. [PMID: 33043029 PMCID: PMC7522218 DOI: 10.3389/fmed.2020.00529] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/27/2020] [Indexed: 12/14/2022] Open
Abstract
Imaging biomarkers of Alzheimer's disease (AD) that are able to detect molecular changes in vivo and transgenic animal models mimicking AD pathologies are essential for the evaluation of new therapeutic strategies. Positron-emission tomography (PET) using either 18F-Fluorodeoxyglucose (18F-FDG) or amyloid-tracers is a well-established, non-invasive tool in the clinical diagnostics of AD assessing two major pathological hallmarks. 18F-FDG-PET is able to detect early changes in cerebral glucose metabolism and amyloid-PET shows cerebral amyloid load. However, the suitability of 18F-FDG- and amyloid-PET in the widely used 5XFAD mouse model of AD is unclear as only a few studies on the use of PET biomarkers are available showing some conflicting results. The aim of this study was the evaluation of 18F-FDG-PET and amyloid-PET in 5XFAD mice in comparison to neurological deficits and neuropathological changes. Seven- and 12-month-old male 5XFAD mice showed a significant reduction in brain glucose metabolism in 18F-FDG-PET and amyloid-PET with 18F-Florbetaben demonstrated an increased cerebral amyloid deposition (n = 4-6 per group). Deficits in spatial reference memory were detected in 12-month-old 5XFAD mice in the Morris Water Maze (n = 10-12 per group). Furthermore, an increased plaque load and gliosis could be proven immunohistochemically in 5XFAD mice (n = 4-6 per group). PET biomarkers 18F-FDG and 18F-Florbetaben detected cerebral hypometabolism and increased plaque load even before the onset of severe memory deficits. Therefore, the 5XFAD mouse model of AD is well-suited for in vivo monitoring of AD pathologies and longitudinal testing of new therapeutic approaches.
Collapse
Affiliation(s)
- Timon N Franke
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Caroline Irwin
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Thomas A Bayer
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Winfried Brenner
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Nicola Beindorff
- Berlin Experimental Radionuclide Imaging Center (BERIC), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Caroline Bouter
- Department of Nuclear Medicine, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Yvonne Bouter
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| |
Collapse
|
28
|
Hybrid PET/MRI enables high-spatial resolution, quantitative imaging of amyloid plaques in an Alzheimer's disease mouse model. Sci Rep 2020; 10:10379. [PMID: 32587315 PMCID: PMC7316864 DOI: 10.1038/s41598-020-67284-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/07/2020] [Indexed: 01/08/2023] Open
Abstract
The emergence of PET probes for amyloid plaques and neurofibrillary tangles, hallmarks of Alzheimer disease (AD), enables monitoring of pathology in AD mouse models. However, small-animal PET imaging is limited by coarse spatial resolution. We have installed a custom-fabricated PET insert into our small-animal MRI instrument and used PET/MRI hybrid imaging to define regions of amyloid vulnerability in 5xFAD mice. We compared fluorine-18 [18F]-Florbetapir uptake in the 5xFAD brain by dedicated small-animal PET/MRI and PET/CT to validate the quantitative measurement of PET/MRI. Next, we used PET/MRI to define uptake in six brain regions. As expected, uptake was comparable to wild-type in the cerebellum and elevated in the cortex and hippocampus, regions implicated in AD. Interestingly, uptake was highest in the thalamus, a region often overlooked in AD studies. Development of small-animal PET/MRI enables tracking of brain region-specific pathology in mouse models, which may prove invaluable to understanding AD progression and therapeutic development.
Collapse
|
29
|
Šála M, Hollinger KR, Thomas AG, Dash RP, Tallon C, Veeravalli V, Lovell L, Kögler M, Hřebabecký H, Procházková E, Nešuta O, Donoghue A, Lam J, Rais R, Rojas C, Slusher BS, Nencka R. Novel Human Neutral Sphingomyelinase 2 Inhibitors as Potential Therapeutics for Alzheimer's Disease. J Med Chem 2020; 63:6028-6056. [PMID: 32298582 PMCID: PMC8025741 DOI: 10.1021/acs.jmedchem.0c00278] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Neutral sphingomyelinase 2 (nSMase2) catalyzes the cleavage of sphingomyelin to phosphorylcholine and ceramide, an essential step in the formation and release of exosomes from cells that is critical for intracellular communication. Chronic increase of brain nSMase2 activity and related exosome release have been implicated in various pathological processes, including the progression of Alzheimer's disease (AD), making nSMase2 a viable therapeutic target. Recently, we identified phenyl (R)-(1-(3-(3,4-dimethoxyphenyl)-2,6-dimethylimidazo[1,2-b]pyridazin-8-yl)pyrrolidin-3-yl)carbamate 1 (PDDC), the first nSMase2 inhibitor that possesses both favorable pharmacodynamics and pharmacokinetic (PK) parameters, including substantial oral bioavailability, brain penetration, and significant inhibition of exosome release from the brain in vivo. Herein we demonstrate the efficacy of 1 (PDDC) in a mouse model of AD and detail extensive structure-activity relationship (SAR) studies with 70 analogues, unveiling several that exert similar or higher activity against nSMase2 with favorable pharmacokinetic properties.
Collapse
Affiliation(s)
- Michal Šála
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nám. 2, 166 10 Prague 6, Czech Republic
| | | | | | | | | | | | | | - Martin Kögler
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nám. 2, 166 10 Prague 6, Czech Republic
| | - Hubert Hřebabecký
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nám. 2, 166 10 Prague 6, Czech Republic
| | - Eliška Procházková
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nám. 2, 166 10 Prague 6, Czech Republic
| | - Ondřej Nešuta
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nám. 2, 166 10 Prague 6, Czech Republic
| | | | | | | | | | | | - Radim Nencka
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nám. 2, 166 10 Prague 6, Czech Republic
| |
Collapse
|
30
|
Barron AM, Ji B, Fujinaga M, Zhang MR, Suhara T, Sahara N, Aoki I, Tsukada H, Higuchi M. In vivo positron emission tomography imaging of mitochondrial abnormalities in a mouse model of tauopathy. Neurobiol Aging 2020; 94:140-148. [PMID: 32623260 DOI: 10.1016/j.neurobiolaging.2020.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 04/05/2020] [Accepted: 05/04/2020] [Indexed: 01/06/2023]
Abstract
Damaged mitochondria may be one of the earliest manifestations of Alzheimer's disease. Because oxidative phosphorylation is a primary source of neuronal energy, unlike glycolysis-dependent energy production in inflamed glia, mitochondrial respiration could provide a selective biomarker of neuronal deterioration in Alzheimer's disease. Here we used a recently developed positron emission tomography (PET) probe targeting mitochondrial complex I (MC-I), 18F-BCPP-EF, to non-invasively visualize mitochondrial abnormalities in the brains of tau transgenic mice (rTg4510). Tauopathy and neuroinflammation were visualized by PET using a tau probe 11C-PBB3 and a translocator protein probe, 18F-FEBMP, respectively. A marked reduction in 18F-BCPP-EF uptake was observed in hippocampal and forebrain regions of tau transgenic mice, colocalizing with regions of tauopathy, neuronal damage, and neuroinflammation. MC-I signals were highly correlated with atrophy assayed by magnetic resonance imaging, but negatively associated with inflammatory signals, indicating that neuronal metabolic signals measured by MC-I PET were robust to inflammatory interference. MC-I may be a useful imaging biomarker to detect neuronal damage and metabolic changes with minimal interference from concomitant glial hypermetabolism.
Collapse
Affiliation(s)
- Anna M Barron
- Department of Functional Brain Imaging, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan; Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore.
| | - Bin Ji
- Department of Functional Brain Imaging, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Masayuki Fujinaga
- Department of Functional Brain Imaging, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Ming-Rong Zhang
- Department of Functional Brain Imaging, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Tetsuya Suhara
- Department of Functional Brain Imaging, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Naruhiko Sahara
- Department of Functional Brain Imaging, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Ichio Aoki
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Hideo Tsukada
- Central Research Laboratory, Hamamatsu Photonics K.K., Shizuoka, Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan.
| |
Collapse
|
31
|
Bouter C, Bouter Y. 18F-FDG-PET in Mouse Models of Alzheimer's Disease. Front Med (Lausanne) 2019; 6:71. [PMID: 31058151 PMCID: PMC6482246 DOI: 10.3389/fmed.2019.00071] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/21/2019] [Indexed: 01/08/2023] Open
Abstract
Suitable animal models and in vivo biomarkers are essential for development and evaluation of new therapeutic strategies in Alzheimer's disease (AD). 18F-Fluorodeoxyglucose (18F-FDG)-positron-emission tomography (PET) is an imaging biomarker that allows the assessment of cerebral glucose metabolism in vivo. While 18F-FDG-PET/CT is an established tool in the evaluation of AD patients, its role in preclinical studies with AD mouse models remains unclear. Here, we want to review available studies on 18F-FDG-PET/CT in AD mouse models in order to evaluate the method and its impact in preclinical AD research. Only a limited number of studies using 18F-FDG-PET in AD mice were carried out so far showing contradictory findings in cerebral FDG uptake. Methodological differences as well as underlying pathological features of used mouse models seem to be accountable for those varying results. However, 18F-FDG-PET can be a valuable tool in longitudinal in vivo therapy monitoring with a lot of potential for future studies.
Collapse
Affiliation(s)
- Caroline Bouter
- Department of Nuclear Medicine, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Yvonne Bouter
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| |
Collapse
|
32
|
Effect of genotype and age on cerebral [ 18F]FDG uptake varies between transgenic APP Swe-PS1 dE9 and Tg2576 mouse models of Alzheimer's disease. Sci Rep 2019; 9:5700. [PMID: 30952945 PMCID: PMC6450965 DOI: 10.1038/s41598-019-42074-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 03/22/2019] [Indexed: 12/17/2022] Open
Abstract
Back-translation of clinical imaging biomarkers of Alzheimer’s disease (AD), such as alterations in cerebral glucose metabolism detected by [18F]FDG positron emission tomography (PET), would be valuable for preclinical studies evaluating new disease-modifying drugs for AD. However, previous confounding results have been difficult to interpret due to differences in mouse models and imaging protocols between studies. We used an equivalent study design and [18F]FDG µPET imaging protocol to compare changes in cerebral glucose metabolism in commercial transgenic APPSwe-PS1dE9 (n = 12), Tg2576 (n = 15), and wild-type mice (n = 15 and 9). Dynamic [18F]FDG scans were performed in young (6 months) and aged (12 or 17 months) mice and the results verified by ex vivo methods (i.e., tissue counting, digital autoradiography, and beta-amyloid and Iba-1 immunohistochemistry). [18F]FDG uptake exhibited significant regional differences between genotypes (TG < WT) and ages (6 months <12 months) in the APPSwe-PS1dE9 model, whereas similar differences were not present in Tg2576 mice. In both models, only weak correlations were detected between regional beta-amyloid deposition or microgliosis and [18F]FDG uptake. By using equivalent methodology, this study demonstrated differences in cerebral glucose metabolism dysfunction detected with [18F]FDG PET between two widely used commercial AD mouse models.
Collapse
|
33
|
Adlimoghaddam A, Snow WM, Stortz G, Perez C, Djordjevic J, Goertzen AL, Ko JH, Albensi BC. Regional hypometabolism in the 3xTg mouse model of Alzheimer's disease. Neurobiol Dis 2019; 127:264-277. [PMID: 30878533 DOI: 10.1016/j.nbd.2019.03.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 02/22/2019] [Accepted: 03/12/2019] [Indexed: 12/28/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive age-related neurodegenerative disease. Although neurofibrillary tangles and amyloid beta are classic hallmarks of AD, the earliest deficits in AD progression may be caused by unknown factors. One suspected factor has to do with brain energy metabolism. To investigate this factor, brain metabolic activity in 3xTg-AD mice and age-matched controls were measured with FDG-PET. Significant hypometabolic changes (p < .01) in brain metabolism were detected in the cortical piriform and insular regions of AD brains relative to controls. These regions are associated with olfaction, which is a potential clinical marker for AD progression as well as neurogenesis. The activity of the terminal component of the mitochondrial respiratory chain (complex IV) and the expression of complex I-V were significantly decreased (p < .05), suggesting that impaired metabolic activity coupled with impaired oxidative phosphorylation leads to decreased mitochondrial bioenergetics and subsequent Neurodegeneration. Although there is an association between neuroinflammatory pathological markers (microglial) and hypometabolism in AD, there was no association found between neuropathological (Aβ, tau, and astrocytes) and functional changes in AD sensitive brain regions, also suggesting that brain hypometabolism occurs prior to AD pathology. Therefore, targeting metabolic mechanisms in cortical piriform and insular regions at early stages may be a promising approach for preventing, slowing, and/or blocking the onset of AD and preserving neurogenesis.
Collapse
Affiliation(s)
- Aida Adlimoghaddam
- St. Boniface Hospital Research, Canada; Dept. of Pharmacology & Therapeutics, University of Manitoba, Canada.
| | | | | | - Claudia Perez
- St. Boniface Hospital Research, Canada; Dept. of Pharmacology & Therapeutics, University of Manitoba, Canada
| | - Jelena Djordjevic
- St. Boniface Hospital Research, Canada; Dept. of Pharmacology & Therapeutics, University of Manitoba, Canada
| | | | - Ji Hyun Ko
- Dept. of Human Anatomy and Cell Science, University of Manitoba, Canada; Health Sciences Centre, Canada
| | - Benedict C Albensi
- St. Boniface Hospital Research, Canada; Dept. of Pharmacology & Therapeutics, University of Manitoba, Canada.
| |
Collapse
|
34
|
Ostapchenko VG, Snir J, Suchy M, Fan J, Cobb MR, Chronik BA, Kovacs M, Prado VF, Hudson RHE, Pasternak SH, Prado MAM, Bartha R. Detection of Active Caspase-3 in Mouse Models of Stroke and Alzheimer's Disease with a Novel Dual Positron Emission Tomography/Fluorescent Tracer [ 68Ga]Ga-TC3-OGDOTA. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2019:6403274. [PMID: 30755766 PMCID: PMC6348924 DOI: 10.1155/2019/6403274] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/20/2018] [Accepted: 11/19/2018] [Indexed: 01/22/2023]
Abstract
Apoptosis is a feature of stroke and Alzheimer's disease (AD), yet there is no accepted method to detect or follow apoptosis in the brain in vivo. We developed a bifunctional tracer [68Ga]Ga-TC3-OGDOTA containing a cell-penetrating peptide separated from fluorescent Oregon Green and 68Ga-bound labels by the caspase-3 recognition peptide DEVD. We hypothesized that this design would allow [68Ga]Ga-TC3-OGDOTA to accumulate in apoptotic cells. In vitro, Ga-TC3-OGDOTA labeled apoptotic neurons following exposure to camptothecin, oxygen-glucose deprivation, and β-amyloid oligomers. In vivo, PET showed accumulation of [68Ga]Ga-TC3-OGDOTA in the brain of mouse models of stroke or AD. Optical clearing revealed colocalization of [68Ga]Ga-TC3-OGDOTA and cleaved caspase-3 in brain cells. In stroke, [68Ga]Ga-TC3-OGDOTA accumulated in neurons in the penumbra area, whereas in AD mice [68Ga]Ga-TC3-OGDOTA was found in single cells in the forebrain and diffusely around amyloid plaques. In summary, this bifunctional tracer is selectively associated with apoptotic cells in vitro and in vivo in brain disease models and represents a novel tool for apoptosis detection that can be used in neurodegenerative diseases.
Collapse
Affiliation(s)
- Valeriy G. Ostapchenko
- Robarts Research Institute, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - Jonatan Snir
- Robarts Research Institute, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Department of Medical Biophysics, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - Mojmir Suchy
- Robarts Research Institute, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Department of Chemistry, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - Jue Fan
- Robarts Research Institute, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - M. Rebecca Cobb
- Robarts Research Institute, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Neuroscience Program, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - Blaine A. Chronik
- Department of Medical Biophysics, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Department of Physics and Astronomy, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - Michael Kovacs
- Department of Medical Biophysics, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Lawson Health Research Institute, 268 Grosvenor Street, London, ON, Canada N6A 4V2
| | - Vania F. Prado
- Robarts Research Institute, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Department of Physiology and Pharmacology, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Department of Anatomy and Cell Biology, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - Robert H. E. Hudson
- Department of Chemistry, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - Stephen H. Pasternak
- Robarts Research Institute, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Department of Clinical Neurological Sciences, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - Marco A. M. Prado
- Robarts Research Institute, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Department of Physiology and Pharmacology, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Department of Anatomy and Cell Biology, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| | - Robert Bartha
- Robarts Research Institute, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
- Department of Medical Biophysics, University of Western Ontario, 1151 Richmond Street, London, ON, Canada N6A 5B7
| |
Collapse
|
35
|
Bouter C, Henniges P, Franke TN, Irwin C, Sahlmann CO, Sichler ME, Beindorff N, Bayer TA, Bouter Y. 18F-FDG-PET Detects Drastic Changes in Brain Metabolism in the Tg4-42 Model of Alzheimer's Disease. Front Aging Neurosci 2019; 10:425. [PMID: 30670962 PMCID: PMC6333025 DOI: 10.3389/fnagi.2018.00425] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/07/2018] [Indexed: 12/19/2022] Open
Abstract
The evaluation of new therapeutic strategies in Alzheimer’s disease (AD) relies heavily on in vivo imaging and suitable animal models that mimic the pathological changes seen in patients. 18F-Fluorodeoxyglucose (18F-FDG)-positron-emission tomography (PET) is a well-established non-invasive imaging tool for monitoring changes in cerebral brain glucose metabolism in vivo. 18F-FDG-PET is used as a functional biomarker for AD as patients show an early and progressive reduction of cerebral glucose metabolism. However, earlier studies in preclinical models of AD showed conflicting results. The aim of this study was the evaluation of cerebral glucose metabolism in the Tg4–42 mouse model of AD using 18F-FDG-PET/magnetic resonance imaging (MRI). Tg4–42 mice show an age-dependent reduction in glucose metabolism together with severe neuron loss and memory deficits. Similar to AD patients early decrease in 18F-FDG uptake was already detected in young (3 months) Tg4–42 mice. The altered glucose metabolism coupled with age- and disease related cognitive decline of Tg4–42 mice make it a well-suited model for preclinical testing of AD-relevant therapeutics.
Collapse
Affiliation(s)
- Caroline Bouter
- Department of Nuclear Medicine, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Philipp Henniges
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Timon N Franke
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Caroline Irwin
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Carsten Oliver Sahlmann
- Department of Nuclear Medicine, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Marius E Sichler
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Nicola Beindorff
- Berlin Experimental Radionuclide Imaging Center (BERIC), Charité-University Medicine Berlin, Berlin, Germany
| | - Thomas A Bayer
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| | - Yvonne Bouter
- Division of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), Georg-August-University, Göttingen, Germany
| |
Collapse
|
36
|
Son HJ, Jeong YJ, Yoon HJ, Lee SY, Choi GE, Park JA, Kim MH, Lee KC, Lee YJ, Kim MK, Cho K, Kang DY. Assessment of brain beta-amyloid deposition in transgenic mouse models of Alzheimer's disease with PET imaging agents 18F-flutemetamol and 18F-florbetaben. BMC Neurosci 2018; 19:45. [PMID: 30053803 PMCID: PMC6063010 DOI: 10.1186/s12868-018-0447-7] [Citation(s) in RCA: 9] [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: 02/09/2018] [Accepted: 07/23/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Although amyloid beta (Aβ) imaging is widely used for diagnosing and monitoring Alzheimer's disease in clinical fields, paralleling comparison between 18F-flutemetamol and 18F-florbetaben was rarely attempted in AD mouse model. We performed a comparison of Aβ PET images between 18F-flutemetamol and 18F-florbetaben in a recently developed APPswe mouse model, C57BL/6-Tg (NSE-hAPPsw) Korl. RESULTS After an injection (0.23 mCi) of 18F-flutemetamol and 18F-florbetaben at a time interval of 2-3 days, we compared group difference of SUVR and kinetic parameters between the AD (n = 7) and control (n = 7) mice, as well as between 18F-flutemetamol and 18F-florbetaben image. In addition, bio-distribution and histopathology were conducted. With visual image and VOI-based SUVR analysis, the AD group presented more prominent uptake than did the control group in both the 18F-florbetaben and 18F-flutemetamol images. With kinetic analysis, the 18F-florbetaben images showed differences in K1 and k4 between the AD and control groups, although 18F-flutemetamol images did not show significant difference. 18F-florbetaben images showed more prominent cortical uptake and matched well to the thioflavin S staining images than did the 18F-flutemetamol image. In contrast, 18F-flutemetamol images presented higher K1, k4, K1/k2 values than those of 18F-florbetaben images. Also, 18F-flutemetamol images presented prominent uptake in the bowel and bladder, consistent with higher bio-distribution in kidney, lung, blood and heart. CONCLUSIONS Compared with 18F-flutemetamol images, 18F-florbetaben images showed prominent visual uptake intensity, SUVR, and higher correlations with the pathology. In contrast, 18F-flutemetamol was more actively metabolized than was 18F-florbetaben (Son et al. in J Nucl Med 58(Suppl 1):S278, 2017].
Collapse
Affiliation(s)
- Hye Joo Son
- Department of Nuclear Medicine, Dong-A University Medical Center, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan, 602-812 Korea
| | - Young Jin Jeong
- Department of Nuclear Medicine, Dong-A University Medical Center, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan, 602-812 Korea
| | - Hyun Jin Yoon
- Department of Nuclear Medicine, Dong-A University Medical Center, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan, 602-812 Korea
| | - Sang Yoon Lee
- Department of Nuclear Medicine, Dong-A University Medical Center, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan, 602-812 Korea
| | - Go-Eun Choi
- Institute of Convergence Bio-Health, Dong-A University, Busan, Korea
| | - Ji-Ae Park
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Min Hwan Kim
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Kyo Chul Lee
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Yong Jin Lee
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Mun Ki Kim
- Pohang Center of Evolution of Biomaterials, Pohang Technopark, Pohang, Korea
| | - Kook Cho
- Institute of Convergence Bio-Health, Dong-A University, Busan, Korea
| | - Do-Young Kang
- Department of Nuclear Medicine, Dong-A University Medical Center, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan, 602-812 Korea
- Institute of Convergence Bio-Health, Dong-A University, Busan, Korea
| |
Collapse
|
37
|
Tolomeo D, Micotti E, Serra SC, Chappell M, Snellman A, Forloni G. Chemical exchange saturation transfer MRI shows low cerebral 2-deoxy-D-glucose uptake in a model of Alzheimer's Disease. Sci Rep 2018; 8:9576. [PMID: 29934551 PMCID: PMC6015016 DOI: 10.1038/s41598-018-27839-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 06/11/2018] [Indexed: 12/17/2022] Open
Abstract
Glucose is the central nervous system's only energy source. Imaging techniques capable to detect pathological alterations of the brain metabolism are useful in different diagnostic processes. Such techniques are also beneficial for assessing the evaluation efficacy of therapies in pre-clinical and clinical stages of diseases. Chemical exchange saturation transfer (CEST) magnetic resonance imaging (MRI) is a possible alternative to positron emission tomography (PET) imaging that has been widely explored in cancer research in humans and animal models. We propose that pathological alterations in brain 2-deoxy-D-glucose (2DG) uptake, typical of neurodegenerative diseases, can be detected with CEST MRI. Transgenic mice overexpressing a mutated form of amyloid precusrsor protein (APP23), a model of Alzheimer's disease, analyzed with CEST MRI showed a clear reduction of 2DG uptake in different brain regions. This was reminiscent of the cerebral condition observed in Alzheimer's patients. The results indicate the feasibility of CEST for analyzing the brain metabolic state, with better image resolution than PET in experimental models.
Collapse
Affiliation(s)
- Daniele Tolomeo
- Laboratory of Biology of Neurodegenerative Disorders, Department of Neuroscience, IRCCS, Mario Negri Institute for Pharmacological Research, Milan, (MI), Italy
| | - Edoardo Micotti
- Laboratory of Biology of Neurodegenerative Disorders, Department of Neuroscience, IRCCS, Mario Negri Institute for Pharmacological Research, Milan, (MI), Italy
| | | | - Michael Chappell
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, 6396, Oxford, UK
| | - Anniina Snellman
- Medicity Research Laboratory, University of Turku, (Tykistökatu 6, FI-20510), Turku, Finland.,Turku PET Centre, University of Turku, (Kiinamyllynkatu 4-8, FI-20520,), Turku, Finland
| | - Gianluigi Forloni
- Laboratory of Biology of Neurodegenerative Disorders, Department of Neuroscience, IRCCS, Mario Negri Institute for Pharmacological Research, Milan, (MI), Italy.
| |
Collapse
|
38
|
Early Detection of A β Deposition in the 5xFAD Mouse by Amyloid PET. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:5272014. [PMID: 29681782 PMCID: PMC5851318 DOI: 10.1155/2018/5272014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 01/12/2018] [Accepted: 01/29/2018] [Indexed: 11/18/2022]
Abstract
Purpose. 18F-FC119S is a positron emission tomography (PET) tracer for imaging β-amyloid (Aβ) plaques in Alzheimer's disease (AD). The aim of this study is to evaluate the efficacy of 18F-FC119S in quantitating Aβ deposition in a mouse model of early amyloid deposition (5xFAD) by PET. Method. Dynamic 18F-FC119S PET images were obtained in 5xFAD (n = 5) and wild-type (WT) mice (n = 7). The brain PET images were spatially normalized to the M. Mirrione T2-weighted mouse brain MR template, and the volumes of interest were then automatically drawn on the cortex, hippocampus, thalamus, and cerebellum. The specific binding of 18F-FC119S to Aβ was quantified as the distribution volume ratio using Logan graphical analysis with the cerebellum as a reference tissue. The Aβ levels in the brain were also confirmed by immunohistochemical analysis. Result. For the 5xFAD group, radioactivity levels in the cortex, the hippocampus, and the thalamus were higher than those for the WT group. In these regions, specific binding was approximately 1.2-fold higher in 5xFAD mice than in WT. Immunohistochemistry supported these findings; the 5xFAD showed severe Aβ deposition in the cortex and hippocampus in contrast to the WT group. Conclusion. These results demonstrated that 18F-FC119S PET can successfully distinguish Aβ depositions in 5xFAD mice from WT.
Collapse
|
39
|
Waldron AM, Wyffels L, Verhaeghe J, Richardson JC, Schmidt M, Stroobants S, Langlois X, Staelens S. Longitudinal Characterization of [18F]-FDG and [18F]-AV45 Uptake in the Double Transgenic TASTPM Mouse Model. J Alzheimers Dis 2018; 55:1537-1548. [PMID: 27911309 PMCID: PMC5181675 DOI: 10.3233/jad-160760] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We aimed to monitor the timing of amyloid-β deposition in relation to changes in brain function using in vivo imaging with [18F]-AV45 and [18F]-FDG in a mouse model of Alzheimer’s disease. TASTPM transgenic mice and wild-type controls were scanned longitudinally with [18F]-AV45 and [18F]-FDG before (3 months of age) and at multiple time points after the onset of amyloid deposition (6, 9, 12, and 15 months of age). As expected with increasing amyloidosis, TASTPM mice demonstrated progressive age-dependent increases in [18F]-AV45 uptake that were significantly higher than for WT from 9 months onwards and correlated to ex vivo measures of amyloid burden. The metabolism of [18F]-AV45 produces several brain penetrant radiometabolites and normalization to a reference region helps to negate this non-specific binding and improve the sensitivity of [18F]-AV45. The observed trajectory of [18F]-FDG alterations deviated from our proposed hypothesis of gradual decreases with worsening amyloidosis. While [18F]-FDG uptake in TASTPM mice was significantly lower than that of WT at 9 months, reduced [18F]-FDG was not associated with aging in TASTPM mice. Moreover, [18F]-FDG uptake did not correlate to measures of ex vivo amyloid burden. Our findings suggest that while amyloid-β is sufficient to induce hypometabolism, these pathologies are not linked in a dose-dependent manner in TASTPM mice.
Collapse
Affiliation(s)
- Ann-Marie Waldron
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Leonie Wyffels
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium.,Department of Nuclear Medicine, University Hospital Antwerp, Antwerp, Belgium
| | - Jeroen Verhaeghe
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | | | - Mark Schmidt
- Department of Neuroscience, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Sigrid Stroobants
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium.,Department of Nuclear Medicine, University Hospital Antwerp, Antwerp, Belgium
| | - Xavier Langlois
- Department of Neuroscience, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
40
|
Snir JA, Suchy M, Bindseil GA, Kovacs M, Chronik BA, Hudson RH, Pasternak SH, Bartha R. An Aspartyl Cathepsin Targeted PET Agent: Application in an Alzheimer’s Disease Mouse Model. J Alzheimers Dis 2018; 61:1241-1252. [DOI: 10.3233/jad-170115] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Jonatan A. Snir
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | - Mojmir Suchy
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, ON, Canada
- Department of Chemistry, University of Western Ontario, London, ON, Canada
| | - Geron A. Bindseil
- Department of Physics and Astronomy, University of Western Ontario, London, ON, Canada
| | - Michael Kovacs
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
| | - Blaine A. Chronik
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, ON, Canada
- Department of Physics and Astronomy, University of Western Ontario, London, ON, Canada
| | - Robert H.E. Hudson
- Department of Chemistry, University of Western Ontario, London, ON, Canada
| | - Stephen H. Pasternak
- Lawson Health Research Institute, London, ON, Canada
- J. Allyn Taylor Centre for Cell Biology, Molecular Brain Research Group, Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | - Robert Bartha
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, ON, Canada
| |
Collapse
|
41
|
Liu W, Zhuo P, Li L, Jin H, Lin B, Zhang Y, Liang S, Wu J, Huang J, Wang Z, Lin R, Chen L, Tao J. Activation of brain glucose metabolism ameliorating cognitive impairment in APP/PS1 transgenic mice by electroacupuncture. Free Radic Biol Med 2017; 112:174-190. [PMID: 28756309 DOI: 10.1016/j.freeradbiomed.2017.07.024] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 07/24/2017] [Accepted: 07/25/2017] [Indexed: 02/06/2023]
Abstract
An essential feature of Alzheimer's disease (AD) is implicated in brain energy metabolic impairment that is considered underlying pathogenesis of cognitive impairment. Therefore, therapeutic interventions to allay cognitive deficits that target energy metabolism may be an efficacy strategy in AD. In this study, we found that electroacupuncture (EA) at the DU20 acupoint obviously increased glucose metabolism in specific brain regions such as cortex, hippocampus, cingulate gyrus, basal forebrain septum, brain stem, and cerebellum in APP/PS1 transgenic mice by animal 18F-Fluoro-2-deoxy-D-Glucose (18F-FDG)/positron emission tomography (PET) imaging, accompanied by cognitive improvements in the spatial reference learning and memory and memory flexibility and novel object recognition performances. Further evidence shown energy metabolism occurred in neurons or non-neuronal cells of the cortex and hippocampus in terms of the co-location of GLUT3/NeuN and GLUT1/GFAP. Simultaneously, metabolic homeostatic factors were critical for glucose metabolism, including phosphorylated adenosine monophosphate-activated protein kinase (AMPK) and AKT serine/threonine kinase. Furthermore, EA-induced phosphorylated AMPK and AKT inhibited the phosphorylation level of the mammalian target of rapamycin (mTOR) to decrease the accumulation of amyloid-beta (Aβ) in the cortex and hippocampus. These findings are concluded that EA is a potential therapeutic target for delaying memory decline and Aβ deposition of AD. The AMPK and AKT are implicated in the EA-induced cortical and hippocampal energy metabolism, which served as a contributor to improving cognitive function and Aβ deposition in a transgenic mouse model of AD.
Collapse
Affiliation(s)
- Weilin Liu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Peiyuan Zhuo
- Fujian Key Laboratory of Rehabilitation Technology, Fuzhou, Fujian 350122, China
| | - Long Li
- Fujian Key Laboratory of Rehabilitation Technology, Fuzhou, Fujian 350122, China
| | - Hao Jin
- Fujian Key Laboratory of Rehabilitation Technology, Fuzhou, Fujian 350122, China
| | - Bingbing Lin
- Fujian Key Laboratory of Rehabilitation Technology, Fuzhou, Fujian 350122, China
| | - Yingzheng Zhang
- Fujian Key Laboratory of Rehabilitation Technology, Fuzhou, Fujian 350122, China
| | - Shengxiang Liang
- Division of Nuclear Technology and Applications, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Wu
- Fujian Key Laboratory of Rehabilitation Technology, Fuzhou, Fujian 350122, China
| | - Jia Huang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Zhifu Wang
- Fujian Key Laboratory of Rehabilitation Technology, Fuzhou, Fujian 350122, China
| | - Ruhui Lin
- Fujian Key Laboratory of Rehabilitation Technology, Fuzhou, Fujian 350122, China
| | - Lidian Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China.
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China.
| |
Collapse
|
42
|
Neth BJ, Craft S. Insulin Resistance and Alzheimer's Disease: Bioenergetic Linkages. Front Aging Neurosci 2017; 9:345. [PMID: 29163128 PMCID: PMC5671587 DOI: 10.3389/fnagi.2017.00345] [Citation(s) in RCA: 186] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/13/2017] [Indexed: 12/14/2022] Open
Abstract
Metabolic dysfunction is a well-established feature of Alzheimer's disease (AD), evidenced by brain glucose hypometabolism that can be observed potentially decades prior to the development of AD symptoms. Furthermore, there is mounting support for an association between metabolic disease and the development of AD and related dementias. Individuals with insulin resistance, type 2 diabetes mellitus (T2D), hyperlipidemia, obesity, or other metabolic disease may have increased risk for the development of AD and similar conditions, such as vascular dementia. This association may in part be due to the systemic mitochondrial dysfunction that is common to these pathologies. Accumulating evidence suggests that mitochondrial dysfunction is a significant feature of AD and may play a fundamental role in its pathogenesis. In fact, aging itself presents a unique challenge due to inherent mitochondrial dysfunction and prevalence of chronic metabolic disease. Despite the progress made in understanding the pathogenesis of AD and in the development of potential therapies, at present we remain without a disease-modifying treatment. In this review, we will discuss insulin resistance as a contributing factor to the pathogenesis of AD, as well as the metabolic and bioenergetic disruptions linking insulin resistance and AD. We will also focus on potential neuroimaging tools for the study of the metabolic dysfunction commonly seen in AD with hopes of developing therapeutic and preventative targets.
Collapse
Affiliation(s)
- Bryan J Neth
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Suzanne Craft
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
43
|
Mouse models of neurodegenerative disease: preclinical imaging and neurovascular component. Brain Imaging Behav 2017; 12:1160-1196. [PMID: 29075922 DOI: 10.1007/s11682-017-9770-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neurodegenerative diseases represent great challenges for basic science and clinical medicine because of their prevalence, pathologies, lack of mechanism-based treatments, and impacts on individuals. Translational research might contribute to the study of neurodegenerative diseases. The mouse has become a key model for studying disease mechanisms that might recapitulate in part some aspects of the corresponding human diseases. Neurodegenerative disorders are very complicated and multifactorial. This has to be taken in account when testing drugs. Most of the drugs screening in mice are very difficult to be interpretated and often useless. Mouse models could be condiderated a 'pathway models', rather than as models for the whole complicated construct that makes a human disease. Non-invasive in vivo imaging in mice has gained increasing interest in preclinical research in the last years thanks to the availability of high-resolution single-photon emission computed tomography (SPECT), positron emission tomography (PET), high field Magnetic resonance, Optical Imaging scanners and of highly specific contrast agents. Behavioral test are useful tool to characterize different animal models of neurodegenerative pathology. Furthermore, many authors have observed vascular pathological features associated to the different neurodegenerative disorders. Aim of this review is to focus on the different existing animal models of neurodegenerative disorders, describe behavioral tests and preclinical imaging techniques used for diagnose and describe the vascular pathological features associated to these diseases.
Collapse
|
44
|
Takkinen JS, López-Picón FR, Al Majidi R, Eskola O, Krzyczmonik A, Keller T, Löyttyniemi E, Solin O, Rinne JO, Haaparanta-Solin M. Brain energy metabolism and neuroinflammation in ageing APP/PS1-21 mice using longitudinal 18F-FDG and 18F-DPA-714 PET imaging. J Cereb Blood Flow Metab 2017; 37:2870-2882. [PMID: 27834284 PMCID: PMC5536795 DOI: 10.1177/0271678x16677990] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Preclinical animal model studies of brain energy metabolism and neuroinflammation in Alzheimer's disease have produced conflicting results, hampering both the elucidation of the underlying disease mechanism and the development of effective Alzheimer's disease therapies. Here, we aimed to quantify the relationship between brain energy metabolism and neuroinflammation in the APP/PS1-21 transgenic mouse model of Alzheimer's disease using longitudinal in vivo18F-FDG and 18F-DPA-714) PET imaging and ex vivo brain autoradiography. APP/PS1-21 (TG, n = 9) and wild type control mice (WT, n = 9) were studied longitudinally every third month from age 6 to 15 months with 18F-FDG and 18F-DPA-714 with a one-week interval between the scans. Additional TG (n = 52) and WT (n = 29) mice were used for ex vivo studies. In vivo, the 18F-FDG SUVs were lower and the 18F-DPA-714 binding ratios relative to the cerebellum were higher in the TG mouse cortex and hippocampus than in WT mice at age 12 to 15 months ( p < 0.05). The ex vivo cerebellum binding ratios supported the results of the in vivo18F-DPA-714 studies but not the 18F-FDG studies. This longitudinal PET study demonstrated decreased energy metabolism and increased inflammation in the brains of APP/PS1-21 mice compared to WT mice.
Collapse
Affiliation(s)
- Jatta S Takkinen
- 1 MediCity Research Laboratory, University of Turku, Turku, Finland.,2 Turku PET Centre, University of Turku, Turku, Finland
| | - Francisco R López-Picón
- 1 MediCity Research Laboratory, University of Turku, Turku, Finland.,2 Turku PET Centre, University of Turku, Turku, Finland
| | - Rana Al Majidi
- 1 MediCity Research Laboratory, University of Turku, Turku, Finland.,2 Turku PET Centre, University of Turku, Turku, Finland
| | - Olli Eskola
- 2 Turku PET Centre, University of Turku, Turku, Finland
| | | | - Thomas Keller
- 2 Turku PET Centre, University of Turku, Turku, Finland
| | | | - Olof Solin
- 2 Turku PET Centre, University of Turku, Turku, Finland.,4 Turku PET Centre, Åbo Akademi University, Turku, Finland.,5 Department of Chemistry, University of Turku, Turku, Finland
| | - Juha O Rinne
- 6 Turku PET Centre, Turku University Hospital, Turku, Finland.,7 Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland
| | - Merja Haaparanta-Solin
- 1 MediCity Research Laboratory, University of Turku, Turku, Finland.,2 Turku PET Centre, University of Turku, Turku, Finland
| |
Collapse
|
45
|
Contrast-enhanced MR microscopy of amyloid plaques in five mouse models of amyloidosis and in human Alzheimer's disease brains. Sci Rep 2017; 7:4955. [PMID: 28694463 PMCID: PMC5504006 DOI: 10.1038/s41598-017-05285-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 05/25/2017] [Indexed: 11/08/2022] Open
Abstract
Gadolinium (Gd)-stained MRI is based on Gd contrast agent (CA) administration into the brain parenchyma. The strong signal increase induced by Gd CA can be converted into resolution enhancement to record microscopic MR images. Moreover, inhomogeneous distribution of the Gd CA in the brain improves the contrast between different tissues and provides new contrasts in MR images. Gd-stained MRI detects amyloid plaques, one of the microscopic lesions of Alzheimer’s disease (AD), in APPSL/PS1M146L mice or in primates. Numerous transgenic mice with various plaque typologies have been developed to mimic cerebral amyloidosis and comparison of plaque detection between animal models and humans with new imaging methods is a recurrent concern. Here, we investigated detection of amyloid plaques by Gd-stained MRI in five mouse models of amyloidosis (APPSL/PS1M146L, APP/PS1dE9, APP23, APPSwDI, and 3xTg) presenting with compact, diffuse and intracellular plaques as well as in post mortem human-AD brains. The brains were then evaluated by histology to investigate the impact of size, compactness, and iron load of amyloid plaques on their detection by MRI. We show that Gd-stained MRI allows detection of compact amyloid plaques as small as 25 µm, independently of their iron load, in mice as well as in human-AD brains.
Collapse
|
46
|
Neuronal p38α mediates synaptic and cognitive dysfunction in an Alzheimer's mouse model by controlling β-amyloid production. Sci Rep 2017; 7:45306. [PMID: 28361984 PMCID: PMC5374488 DOI: 10.1038/srep45306] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 02/23/2017] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by a severe and progressive neuronal loss leading to cognitive dysfunctions. Previous reports, based on the use of chemical inhibitors, have connected the stress kinase p38α to neuroinflammation, neuronal death and synaptic dysfunction. To explore the specific role of neuronal p38α signalling in the appearance of pathological symptoms, we have generated mice that combine expression of the 5XFAD transgenes to induce AD symptoms with the downregulation of p38α only in neurons (5XFAD/p38α∆-N). We found that the neuronal-specific deletion of p38α improves the memory loss and long-term potentiation impairment induced by 5XFAD transgenes. Furthermore, 5XFAD/p38α∆-N mice display reduced amyloid-β accumulation, improved neurogenesis, and important changes in brain cytokine expression compared with 5XFAD mice. Our results implicate neuronal p38α signalling in the synaptic plasticity dysfunction and memory impairment observed in 5XFAD mice, by regulating both amyloid-β deposition in the brain and the relay of this accumulation to mount an inflammatory response, which leads to the cognitive deficits.
Collapse
|
47
|
Coleman RA, Liang C, Patel R, Ali S, Mukherjee J. Brain and Brown Adipose Tissue Metabolism in Transgenic Tg2576 Mice Models of Alzheimer Disease Assessed Using 18F-FDG PET Imaging. Mol Imaging 2017; 16:1536012117704557. [PMID: 28654383 PMCID: PMC5470140 DOI: 10.1177/1536012117704557] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 03/06/2017] [Accepted: 03/10/2017] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Imaging animal models of Alzheimer disease (AD) is useful for the development of therapeutic drugs and understanding AD. Transgenic Swedish hAPPswe Tg2576 mice are a good model of β-amyloid plaques. We report 18F-fluoro-2-deoxyglucose (18F-FDG) positron emission tomography (PET) imaging of brain and intrascapular brown adipose tissue (IBAT) in transgenic mice 2576 (Tg2576) and wild-type (WT) mice. METHODS Transgenic Tg2576 mice and WT mice, >18 months were injected intraperitonally with ≈ 25 to 30 MBq 18F-FDG while awake. After 60 minutes, they were anesthetized with isoflurane (2.5%) and imaged with Inveon MicroPET. Select mice were killed, imaged ex vivo, and 20 µm sections cut for autoradiography. 18F-FDG uptake in brain and IBAT PET and brain autoradiographs were analyzed. RESULTS Fasting blood glucose levels averaged 120 mg/dL for WT and 100 mg/dL for Tg2576. Compared to WT, Tg2576 mice exhibited a decrease in SUVglc in the various brain regions. Average reductions in the cerebrum regions were as high as -20%, while changes in cerebellum were -3%. Uptake of 18F-FDG in IBAT decreased by -60% in Tg2576 mice and was found to be significant. Intrascapular brown adipose tissue findings in Tg2576 mice are new and not previously reported. Use of blood glucose for PET data analysis and corpus callosum as reference region for autoradiographic analysis were important to detect change in Tg2576 mice. CONCLUSION Our results suggest that 18F-FDG uptake in the Tg2576 mice brain show 18F-FDG deficits only when blood glucose is taken into consideration.
Collapse
Affiliation(s)
- Robert A. Coleman
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA, USA
| | - Christopher Liang
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA, USA
| | - Rima Patel
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA, USA
| | - Sarah Ali
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA, USA
| | - Jogeshwar Mukherjee
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
48
|
Li XY, Men WW, Zhu H, Lei JF, Zuo FX, Wang ZJ, Zhu ZH, Bao XJ, Wang RZ. Age- and Brain Region-Specific Changes of Glucose Metabolic Disorder, Learning, and Memory Dysfunction in Early Alzheimer's Disease Assessed in APP/PS1 Transgenic Mice Using 18F-FDG-PET. Int J Mol Sci 2016; 17:1707. [PMID: 27763550 PMCID: PMC5085739 DOI: 10.3390/ijms17101707] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/01/2016] [Accepted: 10/04/2016] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease (AD) is a leading cause of dementia worldwide, associated with cognitive deficits and brain glucose metabolic alteration. However, the associations of glucose metabolic changes with cognitive dysfunction are less detailed. Here, we examined the brains of APP/presenilin 1 (PS1) transgenic (Tg) mice aged 2, 3.5, 5 and 8 months using 18F-labed fluorodeoxyglucose (18F-FDG) microPET to assess age- and brain region-specific changes of glucose metabolism. FDG uptake was calculated as a relative standardized uptake value (SUVr). Morris water maze (MWM) was used to evaluate learning and memory dysfunction. We showed a glucose utilization increase in multiple brain regions of Tg mice at 2 and 3.5 months but not at 5 and 8 months. Comparisons of SUVrs within brains showed higher glucose utilization than controls in the entorhinal cortex, hippocampus, and frontal cortex of Tg mice at 2 and 3.5 months but in the thalamus and striatum at 3.5, 5 and 8 months. By comparing SUVrs in the entorhinal cortex and hippocampus, Tg mice were distinguished from controls at 2 and 3.5 months. In MWM, Tg mice aged 2 months shared a similar performance to the controls (prodromal-AD). By contrast, Tg mice failed training tests at 3.5 months but failed all MWM tests at 5 and 8 months, suggestive of partial or complete cognitive deficits (symptomatic-AD). Correlation analyses showed that hippocampal SUVrs were significantly correlated with MWM parameters in the symptomatic-AD stage. These data suggest that glucose metabolic disorder occurs before onset of AD signs in APP/PS1 mice with the entorhinal cortex and hippocampus affected first, and that regional FDG uptake increase can be an early biomarker for AD. Furthermore, hippocampal FDG uptake is a possible indicator for progression of Alzheimer's cognition after cognitive decline, at least in animals.
Collapse
Affiliation(s)
- Xue-Yuan Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Wei-Wei Men
- Center for Magnetic Resonance Imaging, Peking University, Beijing 100871, China.
| | - Hua Zhu
- Department of Pathology, Comparative Medical Center, Peking Union Medical College & Institute of Laboratory Animal Science, Chinese Academy of Medical Science, Beijing 100021, China.
| | - Jian-Feng Lei
- Center for Medical Experiments and Testing, Capital Medical University, Beijing 100069, China.
| | - Fu-Xing Zuo
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Zhan-Jing Wang
- Department of Pathology, Comparative Medical Center, Peking Union Medical College & Institute of Laboratory Animal Science, Chinese Academy of Medical Science, Beijing 100021, China.
| | - Zhao-Hui Zhu
- Center for PET imaging, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Xin-Jie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Ren-Zhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
49
|
Barron AM, Tokunaga M, Zhang MR, Ji B, Suhara T, Higuchi M. Assessment of neuroinflammation in a mouse model of obesity and β-amyloidosis using PET. J Neuroinflammation 2016; 13:221. [PMID: 27578213 PMCID: PMC5006247 DOI: 10.1186/s12974-016-0700-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 08/20/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Obesity has been identified as a risk factor for cognitive decline and Alzheimer's disease (AD). The aim of this study was to investigate the effect of obesity on neuroinflammation and cerebral glucose metabolism using PET in a mouse model of β-amyloidosis and determine the relationship between these PET imaging biomarkers, pathogenic changes, and functional outcomes. METHODS Three-month-old C57BL/J6 mice were fed either a standard (control group) or high-fat diet (obese group) for 3 months and intracerebroventricularly infused with vehicle or human beta amyloid 1-42 (Aβ42). We assessed obesity-induced abnormalities in peripheral metabolic indices including adiposity, fasting glucose, and glucose tolerance. Brain glucose metabolism was assessed by (18)F-FDG PET, and glial activation was assessed using the translocator protein (TSPO) ligand (11)C-PBR-28. TSPO expression was confirmed by immunohistochemistry of brain sections obtained from scanned mice. The association between inflammatory state and (11)C-PBR-28 PET signals was characterized by examination of the cytokine expression profile in both the serum and hippocampus by antibody array. Learning and memory performance was assessed in the object recognition task, and anxiety-related behavior was assessed in the elevated plus maze. RESULTS Obesity combined with Aβ infusion promoted neuroinflammation and cerebral hypermetabolism, and these signals were significant predictors of learning and memory performance in the object recognition task. In vivo TSPO signals were associated with inflammatory markers including CXCL1, CXCL2, CXCL12, CCL3, CCL5, TIMP-1, G-CSF, sICAM-1, and IL-1ra. CONCLUSIONS In vivo cerebral metabolism and TSPO signals indicate that obesity can accelerate amyloid-induced inflammation and associated cognitive decline.
Collapse
Affiliation(s)
- Anna M. Barron
- Department of Functional Brain Imaging Research, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555 Japan
- Neurobiology of Aging and Disease Laboratory, Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921 Singapore
| | - Masaki Tokunaga
- Department of Functional Brain Imaging Research, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555 Japan
| | - Ming-Rong Zhang
- Department of Radiopharmaceutics Development, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555 Japan
| | - Bin Ji
- Department of Functional Brain Imaging Research, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555 Japan
| | - Tetsuya Suhara
- Department of Functional Brain Imaging Research, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555 Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging Research, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555 Japan
| |
Collapse
|
50
|
Griñán-Ferré C, Sarroca S, Ivanova A, Puigoriol-Illamola D, Aguado F, Camins A, Sanfeliu C, Pallàs M. Epigenetic mechanisms underlying cognitive impairment and Alzheimer disease hallmarks in 5XFAD mice. Aging (Albany NY) 2016; 8:664-84. [PMID: 27013617 PMCID: PMC4925821 DOI: 10.18632/aging.100906] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 01/23/2016] [Indexed: 12/27/2022]
Abstract
5XFAD is an early-onset mouse transgenic model of Alzheimer disease (AD). Up to now there are no studies that focus on the epigenetic changes produced as a result of Aβ-42 accumulation and the possible involvement in the different expression of related AD-genes. Under several behavioral and cognition test, we found impairment in memory and psychoemotional changes in female 5XFAD mice in reference to wild type that worsens with age. Cognitive changes correlated with alterations on protein level analysis and gene expression of markers related with tau aberrant phosphorylation, amyloidogenic pathway (APP, BACE1), Oxidative Stress (iNOS, Aldh2) and inflammation (astrogliosis, TNF-α and IL-6); no changes were found in non-amyloidogenic pathway indicators such as ADAM10. Epigenetics changes as higher CpG methylation and transcriptional changes in DNA methyltransferases (DNMTs) family were found. Dnmt1 increases in younger 5XFAD and Dnmt3a and b high levels in the oldest transgenic mice. Similar pattern was found with histone methyltransferases such as Jarid1a andG9a. Histone deacetylase 2 (Hdac2) or Sirt6, both related with cognition and memory, presented a similar pattern. Taken together, these hallmarks presented by the 5XFAD model prompted its use in assessing different potential therapeutic interventions based on epigenetic targets after earlier amyloid deposition.
Collapse
Affiliation(s)
- Christian Griñán-Ferré
- Department of Pharmacology and Therapeutic Chemistry (Pharmacology Section) and Institute of Neuroscience, University of Barcelona, 08028 Barcelona, Spain
| | - Sara Sarroca
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC, and IDIBAPS, 08036 Barcelona, Spain
| | - Aleksandra Ivanova
- Department of Pharmacology and Therapeutic Chemistry (Pharmacology Section) and Institute of Neuroscience, University of Barcelona, 08028 Barcelona, Spain
| | - Dolors Puigoriol-Illamola
- Department of Pharmacology and Therapeutic Chemistry (Pharmacology Section) and Institute of Neuroscience, University of Barcelona, 08028 Barcelona, Spain
| | - Fernando Aguado
- Department of Cellular Biology, University of Barcelona, 08028 Barcelona, Spain
| | - Antoni Camins
- Department of Pharmacology and Therapeutic Chemistry (Pharmacology Section) and Institute of Neuroscience, University of Barcelona, 08028 Barcelona, Spain
| | - Coral Sanfeliu
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC, and IDIBAPS, 08036 Barcelona, Spain
| | - Mercè Pallàs
- Department of Pharmacology and Therapeutic Chemistry (Pharmacology Section) and Institute of Neuroscience, University of Barcelona, 08028 Barcelona, Spain
| |
Collapse
|