1
|
Yao J, Li Z, Zhou Z, Bao A, Wang Z, Wei H, He H. Distinct regional vulnerability to Aβ and iron accumulation in post mortem AD brains. Alzheimers Dement 2024; 20:6984-6997. [PMID: 39175425 PMCID: PMC11485316 DOI: 10.1002/alz.14188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/02/2024] [Accepted: 07/12/2024] [Indexed: 08/24/2024]
Abstract
INTRODUCTION The paramagnetic iron, diamagnetic amyloid beta (Aβ) plaques and their interaction are crucial in Alzheimer's disease (AD) pathogenesis, complicating non-invasive magnetic resonance imaging for prodromal AD detection. METHODS We used a state-of-the-art sub-voxel quantitative susceptibility mapping method to simultaneously measure Aβ and iron levels in post mortem human brains, validated by histology. Further transcriptomic analysis using Allen Human Brain Atlas elucidated the underlying biological processes. RESULTS Regional increased paramagnetic and diamagnetic susceptibility were observed in medial prefrontal, medial parietal, and para-hippocampal cortices associated with iron deposition (R = 0.836, p = 0.003) and Aβ accumulation (R = 0.853, p = 0.002) in AD brains. Higher levels of gene expression relating to cell cycle, post-translational protein modifications, and cellular response to stress were observed. DISCUSSION These findings provide quantitative insights into the variable vulnerability of cortical regions to higher levels of Aβ aggregation, iron overload, and subsequent neurodegeneration, indicating changes preceding clinical symptoms. HIGHLIGHTS The vulnerability of distinct brain regions to amyloid beta (Aβ) and iron accumulation varies. Histological validation was performed on stained sections of ex-vivo human brains. Regional variations in susceptibility were linked to gene expression profiles. Iron and Aβ levels in ex-vivo brains were simultaneously quantified.
Collapse
Affiliation(s)
- Junye Yao
- Center for Brain Imaging Science and TechnologyZhejiang UniversityHangzhouChina
- College of Biomedical Engineering and Instrument ScienceZhejiang UniversityHangzhouChina
| | - Zhenghao Li
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Zihan Zhou
- Center for Brain Imaging Science and TechnologyZhejiang UniversityHangzhouChina
- Stanford University Graduate School of EducationDepartment of RadiologyStanford UniversityStanfordCaliforniaUSA
| | - Aimin Bao
- National Human Brain Bank for Health and DiseaseSchool of Brain Science and Brain MedicineZhejiang UniversityHangzhouChina
| | - Zheng Wang
- School of Psychological and Cognitive SciencesBeijing Key Laboratory of Behavior and Mental HealthIDG/McGovern Institute for Brain ResearchPeking‐Tsinghua Center for Life SciencesPeking UniversityBeijingChina
- School of Biomedical EngineeringHainan UniversityHaikouChina
| | - Hongjiang Wei
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Hongjian He
- Center for Brain Imaging Science and TechnologyZhejiang UniversityHangzhouChina
- School of PhysicsZhejiang UniversityHangzhouChina
- State Key Laboratory of Brain‐Machine IntelligenceZhejiang UniversityHangzhouChina
| |
Collapse
|
2
|
Davies-Jenkins CW, Workman CI, Hupfeld KE, Zöllner HJ, Leoutsakos JM, Kraut MA, Barker PB, Smith GS, Oeltzschner G. Multimodal investigation of neuropathology and neurometabolites in mild cognitive impairment and late-life depression with 11C-PiB beta-amyloid PET and 7T magnetic resonance spectroscopy. Neurobiol Aging 2024; 142:27-40. [PMID: 39111221 DOI: 10.1016/j.neurobiolaging.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 09/02/2024]
Abstract
Positron emission tomography (PET) and magnetic resonance spectroscopy (1H-MRS) are complementary techniques that can be applied to study how proteinopathy and neurometabolism relate to cognitive deficits in preclinical stages of Alzheimer's disease (AD)-mild cognitive impairment (MCI) and late-life depression (LLD). We acquired beta-amyloid (Aβ) PET and 7 T 1H-MRS measures of GABA, glutamate, glutathione, N-acetylaspartate, N-acetylaspartylglutamate, myo-inositol, choline, and lactate in the anterior and posterior cingulate cortices (ACC, PCC) in 13 MCI and 9 LLD patients, and 13 controls. We used linear regression to examine associations between metabolites, Aβ, and cognitive scores, and whether metabolites and Aβ explained cognitive scores better than Aβ alone. In the ACC, higher Aβ was associated with lower GABA in controls but not MCI or LLD patients, but results depended upon MRS data quality control criteria. Greater variance in California Verbal Learning Test scores was better explained by a model that combined ACC glutamate and Aβ deposition than by models that only included one of these variables. These findings identify preliminary associations between Aβ, neurometabolites, and cognition.
Collapse
Affiliation(s)
- Christopher W Davies-Jenkins
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Clifford I Workman
- Division of Geriatric Psychiatry and Neuropsychiatry, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kathleen E Hupfeld
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Helge J Zöllner
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Jeannie-Marie Leoutsakos
- Division of Geriatric Psychiatry and Neuropsychiatry, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Michael A Kraut
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter B Barker
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Gwenn S Smith
- Division of Geriatric Psychiatry and Neuropsychiatry, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Georg Oeltzschner
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| |
Collapse
|
3
|
Norambuena A, Sagar VK, Wang Z, Raut P, Feng Z, Wallrabe H, Pardo E, Kim T, Alam SR, Hu S, Periasamy A, Bloom GS. Disrupted mitochondrial response to nutrients is a presymptomatic event in the cortex of the APP SAA knock-in mouse model of Alzheimer's disease. Alzheimers Dement 2024; 20:6844-6859. [PMID: 39171353 PMCID: PMC11485302 DOI: 10.1002/alz.14144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/13/2024] [Accepted: 07/01/2024] [Indexed: 08/23/2024]
Abstract
INTRODUCTION Reduced brain energy metabolism, mammalian target of rapamycin (mTOR) dysregulation, and extracellular amyloid beta (Aβ) oligomer (xcAβO) buildup are some well-known Alzheimer's disease (AD) features; how they promote neurodegeneration is poorly understood. We previously reported that xcAβOs inhibit nutrient-induced mitochondrial activity (NiMA) in cultured neurons. We now report NiMA disruption in vivo. METHODS Brain energy metabolism and oxygen consumption were recorded in heterozygous amyloid precursor protein knock-in (APPSAA) mice using two-photon fluorescence lifetime imaging and multiparametric photoacoustic microscopy. RESULTS NiMA is inhibited in APPSAA mice before other defects are detected in these Aβ-producing animals that do not overexpress APP or contain foreign DNA inserts into genomic DNA. Glycogen synthase kinase 3 (GSK3β) signals through mTORC1 to regulate NiMA independently of mitochondrial biogenesis. Inhibition of GSK3β with TWS119 stimulates NiMA in cultured human neurons, and mitochondrial activity and oxygen consumption in APPSAA mice. DISCUSSION NiMA disruption in vivo occurs before plaques, neuroinflammation, and cognitive decline in APPSAA mice, and may represent an early stage in human AD. HIGHLIGHTS Amyloid beta blocks communication between lysosomes and mitochondria in vivo. Nutrient-induced mitochondrial activity (NiMA) is disrupted long before the appearance of Alzheimer's disease (AD) histopathology in heterozygous amyloid precursor protein knock-in (APPSAA/+) mice. NiMA is disrupted long before learning and memory deficits in APPSAA/+ mice. Pharmacological interventions can rescue AD-related NiMA disruption in vivo.
Collapse
Affiliation(s)
- Andrés Norambuena
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Vijay Kumar Sagar
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- W.M. Keck Center for Cellular ImagingUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Zhuoying Wang
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Prakash Raut
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Ziang Feng
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Horst Wallrabe
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- W.M. Keck Center for Cellular ImagingUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Evelyn Pardo
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Taylor Kim
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Shagufta Rehman Alam
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- W.M. Keck Center for Cellular ImagingUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Song Hu
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Ammasi Periasamy
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- W.M. Keck Center for Cellular ImagingUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - George S. Bloom
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Cell BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of NeuroscienceUniversity of VirginiaCharlottesvilleVirginiaUSA
| |
Collapse
|
4
|
Okuyama C, Higashi T, Ishizu K, Oishi N, Kusano K, Ito M, Kagawa S, Okina T, Suzuki N, Hasegawa H, Nagahama Y, Watanabe H, Ono M, Yamauchi H. New objective simple evaluation methods of amyloid PET/CT using whole-brain histogram and Top20%-Map. Ann Nucl Med 2024; 38:763-773. [PMID: 38907835 PMCID: PMC11339116 DOI: 10.1007/s12149-024-01956-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
OBJECTIVE This study aims to assess the utility of newly developed objective methods for the evaluation of intracranial abnormal amyloid deposition using PET/CT histogram without use of cortical ROI analyses. METHODS Twenty-five healthy volunteers (HV) and 38 patients with diagnosed or suspected dementia who had undergone 18F-FPYBF-2 PET/CT were retrospectively included in this study. Out of them, 11C-PiB PET/CT had been also performed in 13 subjects. In addition to the conventional methods, namely visual judgment and quantitative analyses using composed standardized uptake value ratio (comSUVR), the PET images were also evaluated by the following new parameters: the skewness and the mode-to-mean ratio (MMR) obtained from the histogram of the brain parenchyma; Top20%-map highlights the areas with high tracer accumulation occupying 20% volume of the total brain parenchymal on the individual's CT images. We evaluated the utility of the new methods using histogram compared with the visual assessment and comSUVR. The results of these new methods between 18F-FPYBF-2 and 11C-PiB were also compared in 13 subjects. RESULTS In visual analysis, 32, 9, and 22 subjects showed negative, border, and positive results, and composed SUVR in each group were 1.11 ± 0.06, 1.20 ± 0.13, and 1.48 ± 0.18 (p < 0.0001), respectively. Visually positive subjects showed significantly low skewness and high MMR (p < 0.0001), and the Top20%-Map showed the presence or absence of abnormal deposits clearly. In comparison between the two tracers, visual evaluation was all consistent, and the ComSUVR, the skewness, the MMR showed significant good correlation. The Top20%-Maps showed similar pattern. CONCLUSIONS Our new methods using the histogram of the brain parenchymal accumulation are simple and suitable for clinical practice of amyloid PET, and Top20%-Map on the individual's brain CT can be of great help for the visual assessment.
Collapse
Affiliation(s)
- Chio Okuyama
- Clinical Research Center, Shiga General Hospital, Moriyama, Japan.
| | - Tatsuya Higashi
- Clinical Research Center, Shiga General Hospital, Moriyama, Japan.
- Department of Molecular Imaging and Theranostics, National Institute of Quantum Science and Technology, Chiba, Japan.
| | - Koichi Ishizu
- Clinical Research Center, Shiga General Hospital, Moriyama, Japan
- Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Naoya Oishi
- Human Brain Research Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kuninori Kusano
- Clinical Research Center, Shiga General Hospital, Moriyama, Japan
- Department of Radiology, Shiga General Hospital, Moriyama, Japan
| | - Miki Ito
- Clinical Research Center, Shiga General Hospital, Moriyama, Japan
- Department of Radiology, Shiga General Hospital, Moriyama, Japan
| | - Shinya Kagawa
- Clinical Research Center, Shiga General Hospital, Moriyama, Japan
| | - Tomoko Okina
- Department of Neurology, Shiga General Hospital, Moriyama, Japan
| | - Norio Suzuki
- Department of Neurology, Shiga General Hospital, Moriyama, Japan
| | - Hiroshi Hasegawa
- Department of Neurology, Shiga General Hospital, Moriyama, Japan
| | - Yasuhiro Nagahama
- Department of Neurology, Shiga General Hospital, Moriyama, Japan
- Department of Psychiatry and Neurology, Kawasaki Memorial Hospital, Kawasaki, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Fundamental Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Masahiro Ono
- Department of Patho-Fundamental Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Hiroshi Yamauchi
- Clinical Research Center, Shiga General Hospital, Moriyama, Japan
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
5
|
T AR, K K, Paul JS. Unveiling metabolic patterns in dementia: Insights from high-resolution quantitative blood-oxygenation-level-dependent MRI. Med Phys 2024; 51:6002-6019. [PMID: 38888202 DOI: 10.1002/mp.17173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/12/2024] [Accepted: 05/08/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Oxygen extraction fraction (OEF) and deoxyhemoglobin (DoHb) levels reflect variations in cerebral oxygen metabolism in demented patients. PURPOSE Delineating the metabolic profiles evident throughout different phases of dementia necessitates an integrated analysis of OEF and DoHb levels. This is enabled by leveraging high-resolution quantitative blood oxygenation level dependent (qBOLD) analysis of magnitude images obtained from a multi-echo gradient-echo MRI (mGRE) scan performed on a 3.0 Tesla scanner. METHODS Achieving superior spatial resolution in qBOLD necessitates the utilization of an mGRE scan with only four echoes, which in turn limits the number of measurements compared to the parameters within the qBOLD model. Consequently, it becomes imperative to discard non-essential parameters to facilitate further analysis. This process entails transforming the qBOLD model into a format suitable for fitting the log-magnitude difference (L-MDif) profiles of the four echo magnitudes present in each brain voxel. In order to bolster spatial specificity, the log-difference qBOLD model undergoes refinement into a representative form, termed as r-qBOLD, particularly when applied to class-averaged L-MDif signals derived through k-means clustering of L-MDif signals from all brain voxels into a predetermined number of clusters. The agreement between parameters estimated using r-qBOLD for different cluster sizes is validated using Bland-Altman analysis, and the model's goodness-of-fit is evaluated using aχ 2 ${\chi ^2}$ -test. Retrospective MRI data of Alzheimer's disease (AD), mild cognitive impairment (MCI), and non-demented patients without neuropathological disorders, pacemakers, other implants, or psychiatric disorders, who completed a minimum of three visits prior to MRI enrolment, are utilized for the study. RESULTS Utilizing a cohort comprising 30 demented patients aged 65-83 years in stages 4-6 representing mild, moderate, and severe stages according to the clinical dementia rating (CDR), matched with an age-matched non-demented control group of 18 individuals, we conducted joint observations of OEF and DoHb levels estimated using r-qBOLD. The observations elucidate metabolic signatures in dementia based on OEF and DoHb levels in each voxel. Our principal findings highlight the significance of spatial patterns of metabolic profiles (metabolic patterns) within two distinct regimes: OEF levels exceeding the normal range (S1-regime), and OEF levels below the normal range (S2-regime). The S1-regime, accompanied by low DoHb levels, predominantly manifests in fronto-parietal and perivascular regions with increase in dementia severity. Conversely, the S2-regime, accompanied by low DoHb levels, is observed in medial temporal (MTL) regions. Other regions with abnormal metabolic patterns included the orbitofrontal cortex (OFC), medial-orbital prefrontal cortex (MOPFC), hypothalamus, ventro-medial prefrontal cortex (VMPFC), and retrosplenial cortex (RSP). Dysfunction in the OFC and MOPFC indicated cognitive and emotional impairment, while hypothalamic involvement potentially indicated preclinical dementia. Reduced metabolic activity in the RSP suggested early-stage AD related functional abnormalities. CONCLUSIONS Integrated analysis of OEF and DoHb levels using r-qBOLD reveals distinct metabolic signatures across dementia phases, highlighting regions susceptible to neuronal loss, vascular involvement, and preclinical indicators.
Collapse
Affiliation(s)
- Arun Raj T
- Division of Medical Informatics, School of Informatics, Kerala University of Digital Sciences Innovation & Technology (DUK), Trivandrum, Kerala, India
| | - Karthik K
- Department of Neuroimaging & Interventional Radiology, National Institute of Mental Health and Neuro-Sciences (NIMHANS), Bengaluru, Karnataka, India
| | - Joseph Suresh Paul
- Division of Medical Informatics, School of Informatics, Kerala University of Digital Sciences Innovation & Technology (DUK), Trivandrum, Kerala, India
| |
Collapse
|
6
|
Granov R, Vedad S, Wang SH, Durham A, Shah D, Pasinetti GM. The Role of the Neural Exposome as a Novel Strategy to Identify and Mitigate Health Inequities in Alzheimer's Disease and Related Dementias. Mol Neurobiol 2024:10.1007/s12035-024-04339-6. [PMID: 38967905 DOI: 10.1007/s12035-024-04339-6] [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: 12/26/2023] [Accepted: 06/28/2024] [Indexed: 07/06/2024]
Abstract
With the continuous increase of the elderly population, there is an urgency to understand and develop relevant treatments for Alzheimer's disease and related dementias (ADRD). In tandem with this, the prevalence of health inequities continues to rise as disadvantaged communities fail to be included in mainstream research. The neural exposome poses as a relevant mechanistic approach and tool for investigating ADRD onset, progression, and pathology as it accounts for several different factors: exogenous, endogenous, and behavioral. Consequently, through the neural exposome, health inequities can be addressed in ADRD research. In this paper, we address how the neural exposome relates to ADRD by contributing to the discourse through defining how the neural exposome can be developed as a tool in accordance with machine learning. Through this, machine learning can allow for developing a greater insight into the application of transferring and making sense of experimental mouse models exposed to health inequities and potentially relate it to humans. The overall goal moving beyond this paper is to define a multitude of potential factors that can increase the risk of ADRD onset and integrate them to create an interdisciplinary approach to the study of ADRD and subsequently translate the findings to clinical research.
Collapse
Affiliation(s)
- Ravid Granov
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Skyler Vedad
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Shu-Han Wang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Andrea Durham
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Divyash Shah
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Giulio Maria Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA.
- Geriatrics Research, Education and Clinical Center, JJ Peters VA Medical Center, Bronx, NY, 10468, USA.
| |
Collapse
|
7
|
Xie H, Liu Q, Zhou B, Chen X, Guo X, Wang H, Li B, Rominger A, Shi K, Liu C. Unified Noise-aware Network for Low-count PET Denoising with Varying Count Levels. IEEE TRANSACTIONS ON RADIATION AND PLASMA MEDICAL SCIENCES 2024; 8:366-378. [PMID: 39391291 PMCID: PMC11463975 DOI: 10.1109/trpms.2023.3334105] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
As PET imaging is accompanied by substantial radiation exposure and cancer risk, reducing radiation dose in PET scans is an important topic. However, low-count PET scans often suffer from high image noise, which can negatively impact image quality and diagnostic performance. Recent advances in deep learning have shown great potential for recovering underlying signal from noisy counterparts. However, neural networks trained on a specific noise level cannot be easily generalized to other noise levels due to different noise amplitude and variances. To obtain optimal denoised results, we may need to train multiple networks using data with different noise levels. But this approach may be infeasible in reality due to limited data availability. Denoising dynamic PET images presents additional challenge due to tracer decay and continuously changing noise levels across dynamic frames. To address these issues, we propose a Unified Noise-aware Network (UNN) that combines multiple sub-networks with varying denoising power to generate optimal denoised results regardless of the input noise levels. Evaluated using large-scale data from two medical centers with different vendors, presented results showed that the UNN can consistently produce promising denoised results regardless of input noise levels, and demonstrate superior performance over networks trained on single noise level data, especially for extremely low-count data.
Collapse
Affiliation(s)
- Huidong Xie
- Department of Biomedical Engineering, Yale University
| | - Qiong Liu
- Department of Biomedical Engineering, Yale University
| | - Bo Zhou
- Department of Biomedical Engineering, Yale University
| | | | - Xueqi Guo
- Department of Biomedical Engineering, Yale University
| | - Hanzhong Wang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern
- Computer Aided Medical Procedures and Augmented Reality, Institute of Informatics 116, Technical University of Munich, Munich, Germany
| | - Chi Liu
- Department of Biomedical Engineering, Yale University
- Department of Radiology and Biomedical Imaging at Yale University
| |
Collapse
|
8
|
Adebambo K, Ojoh O(C. In Silico Investigation of Novel Compounds as Inhibitors of Acetylcholinesterase Enzyme for the Treatment of Alzheimer's Diseases. Int J Alzheimers Dis 2024; 2024:2988685. [PMID: 38371416 PMCID: PMC10869201 DOI: 10.1155/2024/2988685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/23/2023] [Accepted: 01/17/2024] [Indexed: 02/20/2024] Open
Abstract
Alzheimer's disease (AD) is a "progressive, neurodegenerative disease that occurs when nerve cells in the brain die." There are only 4 drugs approved by the United States Food and Drug Administration (FDA). Three (donepezil, rivastigmine, and galantamine) out of these four drugs are anticholinesterase inhibitors, while the fourth one memantine is an N-methyl-D-aspartate (NMDA) receptor inhibitor. Currently, two immunotherapy drugs that target amyloid protein (donanemab and lecanemab) are being considered for the treatment of Alzheimer's disease at an early stage. All these drug molecules are still not the complete answer to the treatment of Alzheimer's disease. A recent report from the Office of National Statistics showed that AD is the leading cause of death in 2022. Therefore, there is an urgency to develop more drugs that can treat AD. Based on this urgency, we aim to investigate how bioactive and already approved drugs could be repurposed for inhibiting the anticholinesterase enzyme using computational studies. To achieve this, the data science tool-Python coding was compiled on Jupyter Notebook to mine bioactive compounds from the ChEMBL database. The most bioactive compounds obtained were further investigated using Molecular Operating Environment (MOE) software to carry out molecular docking and ligand analysis, and this was followed by molecular dynamics simulation production at 35 ns using GROMACS 2022.4 on Archer 2 machine. The molecular dynamic analysis was carried out using HeroMDanalysis software. Data mining of the ChEMBL database was carried out for lipase inhibitors, and this gave CHEMBL-ID 1240685, a peptide molecule, the most active compound at the time of data mining. Further literature studies gave Zoladex an FDA-approved drug for the treatment of breast cancer as another compound of interest. The in silico studies were carried out against the anticholinesterase enzyme using two FDA-approved drugs donepezil and galantamine as a template for comparing the in silico activities of the repurposed drugs. A very useful receptor for this study was PDB-1DX6, a cocrystallized galantamine inhibitor of acetylcholinesterase enzyme. The molecular docking analysis (using ligand interactions) and molecular dynamic analysis (root mean square deviation (RMSD) and root mean square fluctuation (RMSF)) showed that the two peptide molecules CHEMBL-1240685 and Zoladex gave the best binding energy and stability when compared to the FDA-approved drugs (donepezil and galantamine). Finally, further literature studies revealed that Zoladex affects memory reduction; therefore, it was dropped as a possible repurposed drug. Our research showed that CHEMBL-1240685 is a potential compound that could be investigated for the inhibition of anticholinesterase enzyme and might be another drug molecule that could be used to treat Alzheimer's disease.
Collapse
Affiliation(s)
- Kassim Adebambo
- Department of Clinical Pharmaceutical and Biological Science, University of Hertfordshire, Hatfield, UK
| | | |
Collapse
|
9
|
Norambuena A, Sagar VK, Wang Z, Raut P, Feng Z, Wallrabe H, Pardo E, Kim T, Alam SR, Hu S, Periasamy A, Bloom GS. Disrupted mitochondrial response to nutrients is a presymptomatic event in the cortex of the APP SAA knock-in mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.02.578668. [PMID: 38352486 PMCID: PMC10862844 DOI: 10.1101/2024.02.02.578668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Introduction Reduced brain energy metabolism, mTOR dysregulation, and extracellular amyloid-β oligomer (xcAβO) buildup characterize AD; how they collectively promote neurodegeneration is poorly understood. We previously reported that xcAβOs inhibit N utrient-induced M itochondrial A ctivity (NiMA) in cultured neurons. We now report NiMA disruption in vivo . Methods Brain energy metabolism and oxygen consumption were recorded in APP SAA/+ mice using two-photon fluorescence lifetime imaging and multiparametric photoacoustic microscopy. Results NiMA is inhibited in APP SAA/+ mice before other defects are detected in these amyloid-β-producing animals that do not overexpress APP or contain foreign DNA inserts into genomic DNA. GSK3β signals through mTORC1 to regulate NiMA independently of mitochondrial biogenesis. Inhibition of GSK3β with lithium or TWS119 stimulates NiMA in cultured human neurons, and mitochondrial activity and oxygen consumption in APP SAA mice. Conclusion NiMA disruption in vivo occurs before histopathological changes and cognitive decline in APP SAA mice, and may represent an early stage in human AD.
Collapse
|
10
|
Young AL, Oxtoby NP, Garbarino S, Fox NC, Barkhof F, Schott JM, Alexander DC. Data-driven modelling of neurodegenerative disease progression: thinking outside the black box. Nat Rev Neurosci 2024; 25:111-130. [PMID: 38191721 DOI: 10.1038/s41583-023-00779-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2023] [Indexed: 01/10/2024]
Abstract
Data-driven disease progression models are an emerging set of computational tools that reconstruct disease timelines for long-term chronic diseases, providing unique insights into disease processes and their underlying mechanisms. Such methods combine a priori human knowledge and assumptions with large-scale data processing and parameter estimation to infer long-term disease trajectories from short-term data. In contrast to 'black box' machine learning tools, data-driven disease progression models typically require fewer data and are inherently interpretable, thereby aiding disease understanding in addition to enabling classification, prediction and stratification. In this Review, we place the current landscape of data-driven disease progression models in a general framework and discuss their enhanced utility for constructing a disease timeline compared with wider machine learning tools that construct static disease profiles. We review the insights they have enabled across multiple neurodegenerative diseases, notably Alzheimer disease, for applications such as determining temporal trajectories of disease biomarkers, testing hypotheses about disease mechanisms and uncovering disease subtypes. We outline key areas for technological development and translation to a broader range of neuroscience and non-neuroscience applications. Finally, we discuss potential pathways and barriers to integrating disease progression models into clinical practice and trial settings.
Collapse
Affiliation(s)
- Alexandra L Young
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK.
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Neil P Oxtoby
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK.
| | - Sara Garbarino
- Life Science Computational Laboratory, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Nick C Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Frederik Barkhof
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Jonathan M Schott
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Daniel C Alexander
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| |
Collapse
|
11
|
Valentin-Escalera J, Leclerc M, Calon F. High-Fat Diets in Animal Models of Alzheimer's Disease: How Can Eating Too Much Fat Increase Alzheimer's Disease Risk? J Alzheimers Dis 2024; 97:977-1005. [PMID: 38217592 PMCID: PMC10836579 DOI: 10.3233/jad-230118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2023] [Indexed: 01/15/2024]
Abstract
High dietary intake of saturated fatty acids is a suspected risk factor for neurodegenerative diseases, including Alzheimer's disease (AD). To decipher the causal link behind these associations, high-fat diets (HFD) have been repeatedly investigated in animal models. Preclinical studies allow full control over dietary composition, avoiding ethical concerns in clinical trials. The goal of the present article is to provide a narrative review of reports on HFD in animal models of AD. Eligibility criteria included mouse models of AD fed a HFD defined as > 35% of fat/weight and western diets containing > 1% cholesterol or > 15% sugar. MEDLINE and Embase databases were searched from 1946 to August 2022, and 32 preclinical studies were included in the review. HFD-induced obesity and metabolic disturbances such as insulin resistance and glucose intolerance have been replicated in most studies, but with methodological variability. Most studies have found an aggravating effect of HFD on brain Aβ pathology, whereas tau pathology has been much less studied, and results are more equivocal. While most reports show HFD-induced impairment on cognitive behavior, confounding factors may blur their interpretation. In summary, despite conflicting results, exposing rodents to diets highly enriched in saturated fat induces not only metabolic defects, but also cognitive impairment often accompanied by aggravated neuropathological markers, most notably Aβ burden. Although there are important variations between methods, particularly the lack of diet characterization, these studies collectively suggest that excessive intake of saturated fat should be avoided in order to lower the incidence of AD.
Collapse
Affiliation(s)
- Josue Valentin-Escalera
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada)
| | - Manon Leclerc
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada)
| | - Frédéric Calon
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada)
| |
Collapse
|
12
|
Atef MM, Mostafa YM, Ahmed AAM, El-Sayed NM. Simvastatin attenuates aluminium chloride-induced neurobehavioral impairments through activation of TGF-β1/ SMAD2 and GSK3β/β-catenin signalling pathways. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 102:104220. [PMID: 37454825 DOI: 10.1016/j.etap.2023.104220] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/19/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterised by the presence of β-amyloid plaques and acetylcholine depletion leading to neurobehavioral defects. AD was contributed also with downregulation of TGF-β1/SMAD2 and GSK3β/β-catenin pathways. Simvastatin (SMV) improved memory function experimentally and clinically. Hence, this study aimed to investigate the mechanistic role of SMV against aluminium chloride (AlCl3) induced neurobehavioral impairments. AD was induced by AlCl3 (50 mg/kg) for 6 weeks. Mice received Simvastatin (10 or 20 mg/kg) or Donepezil (3 mg/kg) for 6 weeks after that the histopathological, immunohistochemical and biochemical test were examined. Treatment with SMV improved the memory deterioration induced by AlCl3 with significant recovery of the histopathological changes. This was concomitant with the decrease of AChE and Aβ (1-42). SMV provides its neuroprotective effect through upregulating the protein expression of β-catenin, TGF-β1 and downregulating the expression of GSK3β, TLR4 and p-SMAD2.
Collapse
Affiliation(s)
| | - Yasser M Mostafa
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; Department of Pharmacology & Toxicology, Faculty of Pharmacy, Badr University in Cairo, Egypt
| | - Amal A M Ahmed
- Department of Cytology & Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Norhan M El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt.
| |
Collapse
|
13
|
Garin CM, Dhenain M. Mean amplitude of low frequency fluctuations measured by fMRI at 11.7 T in the aging brain of mouse lemur primate. Sci Rep 2023; 13:7970. [PMID: 37198192 DOI: 10.1038/s41598-023-33482-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/13/2023] [Indexed: 05/19/2023] Open
Abstract
Non-human primates are a critical species for the identification of key biological mechanisms in normal and pathological aging. One of these primates, the mouse lemur, has been widely studied as a model of cerebral aging or Alzheimer's disease. The amplitude of low-frequency fluctuations of blood oxygenation level-dependent (BOLD) can be measured with functional MRI. Within specific frequency bands (e.g. the 0.01-0.1 Hz), these amplitudes were proposed to indirectly reflect neuronal activity as well as glucose metabolism. Here, we first created whole brain maps of the mean amplitude of low frequency fluctuations (mALFF) in young mouse lemurs (mean ± SD: 2.1 ± 0.8 years). Then, we extracted mALFF in old lemurs (mean ± SD: 8.8 ± 1.1 years) to identify age-related changes. A high level of mALFF was detected in the temporal cortex (Brodmann area 20), somatosensory areas (Brodmann area 5), insula (Brodmann areas 13-6) and the parietal cortex (Brodmann area 7) of healthy young mouse lemurs. Aging was associated with alterations of mALFF in somatosensory areas (Brodmann area 5) and the parietal cortex (Brodmann area 7).
Collapse
Affiliation(s)
- Clément M Garin
- UMR 9199, Neurodegenerative Diseases Laboratory, Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, Université Paris-Saclay, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut François Jacob, MIRCen, 18 Route du Panorama, 92265, Fontenay-aux-Roses Cedex, France
| | - Marc Dhenain
- UMR 9199, Neurodegenerative Diseases Laboratory, Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, Université Paris-Saclay, 18 Route du Panorama, 92265, Fontenay-aux-Roses, France.
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut François Jacob, MIRCen, 18 Route du Panorama, 92265, Fontenay-aux-Roses Cedex, France.
| |
Collapse
|
14
|
Lin H, Pan T, Wang M, Ge J, Lu J, Ju Z, Chen K, Zhang H, Guan Y, Zhao Q, Shan B, Nie B, Zuo C, Wu P. Metabolic Asymmetry Relates to Clinical Characteristics and Brain Network Abnormalities in Alzheimer's Disease. J Alzheimers Dis 2023:JAD221258. [PMID: 37182878 DOI: 10.3233/jad-221258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
BACKGROUND Metabolic asymmetry has been observed in Alzheimer's disease (AD), but different studies have inconsistent viewpoints. OBJECTIVE To analyze the asymmetry of cerebral glucose metabolism in AD and investigate its clinical significance and potential metabolic network abnormalities. METHODS Standardized uptake value ratios (SUVRs) were obtained from 18F-FDG positron emission tomography (PET) images of all participants, and the asymmetry indices (AIs) were calculated according to the SUVRs. AD group was divided into left/right-dominant or bilateral symmetric hypometabolism (AD-L/AD-R or AD-BI) when more than half of the AIs of the 20 regions of interest (ROIs) were < -2SD, >2SD, or between±1SD. Differences in clinical features among the three AD groups were compared, and the abnormal network characteristics underlying metabolic asymmetry were explored. RESULTS In AD group, the proportions of AD-L, AD-R, and AD-BI were 28.4%, 17.9%, and 18.5%, respectively. AD-L/AD-R groups had younger age of onset and faster rate of cognitive decline than AD-BI group (p < 0.05). The absolute values of AIs in half of the 20 ROIs became higher at follow-up than at baseline (p < 0.05). Compared with those in AD-BI group, metabolic connection strength of network, global efficiency, cluster coefficient, degree centrality and local efficiency were lower, but shortest path length was longer in AD-L and AD-R groups (p < 0.05). CONCLUSION Asymmetric and symmetric hypometabolism may represent different clinical subtypes of AD, which may provide a clue for future studies on the heterogeneity of AD and help to optimize the design of clinical trials.
Collapse
Affiliation(s)
- Huamei Lin
- Deparment of Nuclear Medicine / PET Center, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Tingting Pan
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High EnergyPhysics, Chinese Academy of Sciences, Beijing, China
| | - Min Wang
- Institute of Biomedical Engineering, School of Communication and Information Engineering, Shanghai University, Shanghai, China
| | - Jingjie Ge
- Deparment of Nuclear Medicine / PET Center, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiaying Lu
- Deparment of Nuclear Medicine / PET Center, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zizhao Ju
- Deparment of Nuclear Medicine / PET Center, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Keliang Chen
- National Center for Neurological Disorders & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Huiwei Zhang
- Deparment of Nuclear Medicine / PET Center, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- Deparment of Nuclear Medicine / PET Center, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Qianhua Zhao
- National Center for Neurological Disorders & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Baoci Shan
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High EnergyPhysics, Chinese Academy of Sciences, Beijing, China
| | - Binbin Nie
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High EnergyPhysics, Chinese Academy of Sciences, Beijing, China
| | - Chuantao Zuo
- Deparment of Nuclear Medicine / PET Center, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ping Wu
- Deparment of Nuclear Medicine / PET Center, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Tada T, Hara K, Fujita N, Ito Y, Yamaguchi H, Ohdake R, Kawabata K, Ogura A, Kato T, Yokoi T, Masuda M, Abe S, Miyao S, Naganawa S, Katsuno M, Watanabe H, Sobue G, Kato K. Comparative examination of the pons and corpus callosum as reference regions for quantitative evaluation in positron emission tomography imaging for Alzheimer's disease using 11C-Pittsburgh Compound-B. Ann Nucl Med 2023:10.1007/s12149-023-01843-y. [PMID: 37160863 DOI: 10.1007/s12149-023-01843-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 04/24/2023] [Indexed: 05/11/2023]
Abstract
OBJECTIVES Standardised uptake value ratio (SUVR) is usually obtained by dividing the SUV of the region of interest (ROI) by that of the cerebellar cortex. Cerebellar cortex is not a valid reference in cases where amyloid β deposition or lesions are present. Only few studies have evaluated the use of other regions as references. We compared the validity of the pons and corpus callosum as reference regions for the quantitative evaluation of brain positron emission tomography (PET) using 11C-PiB compared to the cerebellar cortex. METHODS We retrospectively evaluated data from 86 subjects with or without Alzheimer's disease (AD). All subjects underwent magnetic resonance imaging, PET imaging, and cognitive function testing. For the quantitative analysis, three-dimensional ROIs were automatically placed, and SUV and SUVR were obtained. We compared these values between AD and healthy control (HC) groups. RESULTS SUVR data obtained using the pons and corpus callosum as reference regions strongly correlated with that using the cerebellar cortex. The sensitivity and specificity were high when either the pons or corpus callosum was used as the reference region. However, the SUV values of the corpus callosum were different between AD and HC (p < 0.01). CONCLUSIONS Our data suggest that the pons and corpus callosum might be valid reference regions.
Collapse
Affiliation(s)
- Tomohiro Tada
- Department of Radiological Technology, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8560, Japan
| | - Kazuhiro Hara
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Naotoshi Fujita
- Department of Radiological Technology, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8560, Japan
| | - Yoshinori Ito
- Department of Radiological and Medical Laboratory Sciences, Nagoya University Graduate School of Medicine, 1-1-20 Daiko-Minami, Higashi-Ku, Nagoya, 461-8673, Japan
| | - Hiroshi Yamaguchi
- Nagoya University Radioisotope Research Center Medical Branch, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Reiko Ohdake
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan
| | - Kazuya Kawabata
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
- Department of Neurology, Medical University of Innsbruck, Innrain 52, 6020, Innsbruck, Austria
| | - Aya Ogura
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Toshiyasu Kato
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
- Department of Neurology, Anjo Kosei Hospital, 28 Higashihirokute Anjo-Cho, Anjo, 446-8602, Japan
| | - Takamasa Yokoi
- Department of Neurology, Toyohashi Municipal Hospital, 50 Hachikennishi, Aotake-Cho, Toyohashi, 441-8570, Japan
| | - Michihito Masuda
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
- Department of Neurology, Okazaki City Hospital, 1-3 Gosyoai, Kouryuji-Cho, Okazaki, 444-8553, Japan
| | - Shinji Abe
- Department of Radiological Technology, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8560, Japan
| | - Shinichi Miyao
- Department of Neurology, Meitetsu Hospital, 2-26-11 Sakou, Nishiku, Nagoya, Japan
| | - Shinji Naganawa
- Department of Radiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8560, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
- Department of Clinical Research Education, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8560, Japan
| | - Hirohisa Watanabe
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan
| | - Gen Sobue
- Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Japan
| | - Katsuhiko Kato
- Functional Medical Imaging, Biomedical Imaging Sciences, Division of Advanced Information Health Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, 1-1-20 Daiko-Minami, Higashi-Ku, Nagoya, 461-8673, Japan.
| |
Collapse
|
16
|
Song M, Fan X. Systemic Metabolism and Mitochondria in the Mechanism of Alzheimer's Disease: Finding Potential Therapeutic Targets. Int J Mol Sci 2023; 24:ijms24098398. [PMID: 37176104 PMCID: PMC10179273 DOI: 10.3390/ijms24098398] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/30/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Elderly people over the age of 65 are those most likely to experience Alzheimer's disease (AD), and aging and AD are associated with apparent metabolic alterations. Currently, there is no curative medication against AD and only several drugs have been approved by the FDA, but these drugs can only improve the symptoms of AD. Many preclinical and clinical trials have explored the impact of adjusting the whole-body and intracellular metabolism on the pathogenesis of AD. The most recent evidence suggests that mitochondria initiate an integrated stress response to environmental stress, which is beneficial for healthy aging and neuroprotection. There is also an increasing awareness of the differential risk and potential targeting strategies related to the metabolic level and microbiome. As the main participants in intracellular metabolism, mitochondrial bioenergetics, mitochondrial quality-control mechanisms, and mitochondria-linked inflammatory responses have been regarded as potential therapeutic targets for AD. This review summarizes and highlights these advances.
Collapse
Affiliation(s)
- Meiying Song
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
17
|
Yang Z, Cummings JL, Kinney JW, Cordes D. Accelerated hypometabolism with disease progression associated with faster cognitive decline among amyloid positive patients. Front Neurosci 2023; 17:1151820. [PMID: 37123373 PMCID: PMC10140339 DOI: 10.3389/fnins.2023.1151820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/22/2023] [Indexed: 05/02/2023] Open
Abstract
Objective To evaluate the progression of brain glucose metabolism among participants with biological signature of Alzheimer's disease (AD) and its relevance to cognitive decline. Method We studied 602 amyloid positive individuals who underwent 18F-fluorodeoxyglucose PET (FDG-PET) scan, 18F-AV-45 amyloid PET (AV45-PET) scan, structural MRI scan and neuropsychological examination, including 116 cognitively normal (CN) participants, 314 participants diagnosed as mild cognitive impairment (MCI), and 172 participants diagnosed as AD dementia. The first FDG-PET scan satisfying the inclusion criteria was considered as the baseline scan. Cross-sectional analysis were conducted with the baseline FDG-PET data to compare the regional differences between diagnostic groups after adjusting confounding factors. Among these participants, 229 participants (55 CN, 139 MCI, and 35 AD dementia) had two-year follow-up FDG-PET data available. Regional glucose metabolism was computed and the progression rates of regional glucose metabolism were derived from longitudinal FDG-PET scans. Then the group differences of regional progression rates were examined to assess whether glucose metabolism deficit accelerates or becomes stable with disease progression. The association of cognitive decline rate with baseline regional glucose metabolism, and progression rate in longitudinal data, were evaluated. Results Participants with AD dementia showed substantial glucose metabolism deficit than CN and MCI at left hippocampus, in addition to the traditionally reported frontal and parietal-temporal lobe. More substantial metabolic change was observed with the contrast AD - MCI than the contrast MCI - CN, even after adjusting time duration since cognitive symptom onset. With the longitudinal data, glucose metabolism was observed to decline the most rapidly in the AD dementia group and at a slower rate in MCI. Lower regional glucose metabolism was correlated to faster cognitive decline rate with mild-moderate correlations, and the progression rate was correlated to cognitive decline rate with moderate-large correlations. Discussion and conclusion Hippocampus was identified to experience hypometabolism in AD pathology. Hypometabolism accelerates with disease progression toward AD dementia. FDG-PET, particularly longitudinal scans, could potentially help predict how fast cognition declines and assess the impact of treatment in interventional trials.
Collapse
Affiliation(s)
- Zhengshi Yang
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, United States
- Department of Brain Health, University of Nevada, Las Vegas, Las Vegas, NV, United States
| | - Jeffrey L. Cummings
- Department of Brain Health, University of Nevada, Las Vegas, Las Vegas, NV, United States
- Chambers-Grundy Center for Transformative Neuroscience, Pam Quirk Brain Health and Biomarker Laboratory, Department of Brain Health, School of Integrated Health Sciences, University of Nevada, Las Vegas, Las Vegas, NV, United States
| | - Jefferson W. Kinney
- Department of Brain Health, University of Nevada, Las Vegas, Las Vegas, NV, United States
- Chambers-Grundy Center for Transformative Neuroscience, Pam Quirk Brain Health and Biomarker Laboratory, Department of Brain Health, School of Integrated Health Sciences, University of Nevada, Las Vegas, Las Vegas, NV, United States
| | - Dietmar Cordes
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, United States
- Department of Brain Health, University of Nevada, Las Vegas, Las Vegas, NV, United States
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, United States
| | | |
Collapse
|
18
|
Yu X, Zhang L, Lyu Y, Liu T, Zhu D. SUPERVISED DEEP TREE IN ALZHEIMER'S DISEASE. PROCEEDINGS. IEEE INTERNATIONAL SYMPOSIUM ON BIOMEDICAL IMAGING 2023; 2023:10.1109/isbi53787.2023.10230742. [PMID: 38362508 PMCID: PMC10869122 DOI: 10.1109/isbi53787.2023.10230742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
As a progressive neurodegenerative disorder, the pathological changes of Alzheimer's disease (AD) might begin as much as two decades before the manifestation of clinical symptoms. Since the nature of the irreversible pathology of AD, early diagnosis provides a more tractable way for disease intervention and treatment. Therefore, numerous approaches have been developed for early diagnostic purposes. Although several important biomarkers have been established, most of the existing methods show limitations in describing the continuum of AD progression. However, understanding this continuous development is essential to understand the intrinsic progression mechanism of AD. In this work, we proposed a supervised deep tree model (SDTree) to integrate AD progression and individual prediction. The proposed SDTree method models the progression of AD as a tree embedded in a latent space using nonlinear reversed graph embedding. In this way, the continuum of AD progression is encoded into the locations on the tree structure. The learned tree structure can not only represent the continuum of AD but make predictions for new subjects. We evaluated our method on the classification task and achieved promising results on Alzheimer's Disease Neuroimaging Initiative dataset.
Collapse
Affiliation(s)
- Xiaowei Yu
- Computer Science and Engineering, University of Texas at Arlington, TX, USA
| | - Lu Zhang
- Computer Science and Engineering, University of Texas at Arlington, TX, USA
| | - Yanjun Lyu
- Computer Science and Engineering, University of Texas at Arlington, TX, USA
| | - Tianming Liu
- Computer Science, The University of Georgia, Athens, USA
| | - Dajiang Zhu
- Computer Science and Engineering, University of Texas at Arlington, TX, USA
| |
Collapse
|
19
|
Harris WJ, Asselin MC, Hinz R, Parkes LM, Allan S, Schiessl I, Boutin H, Dickie BR. In vivo methods for imaging blood-brain barrier function and dysfunction. Eur J Nucl Med Mol Imaging 2023; 50:1051-1083. [PMID: 36437425 PMCID: PMC9931809 DOI: 10.1007/s00259-022-05997-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 10/09/2022] [Indexed: 11/29/2022]
Abstract
The blood-brain barrier (BBB) is the interface between the central nervous system and systemic circulation. It tightly regulates what enters and is removed from the brain parenchyma and is fundamental in maintaining brain homeostasis. Increasingly, the BBB is recognised as having a significant role in numerous neurological disorders, ranging from acute disorders (traumatic brain injury, stroke, seizures) to chronic neurodegeneration (Alzheimer's disease, vascular dementia, small vessel disease). Numerous approaches have been developed to study the BBB in vitro, in vivo, and ex vivo. The complex multicellular structure and effects of disease are difficult to recreate accurately in vitro, and functional aspects of the BBB cannot be easily studied ex vivo. As such, the value of in vivo methods to study the intact BBB cannot be overstated. This review discusses the structure and function of the BBB and how these are affected in diseases. It then discusses in depth several established and novel methods for imaging the BBB in vivo, with a focus on MRI, nuclear imaging, and high-resolution intravital fluorescence microscopy.
Collapse
Affiliation(s)
- William James Harris
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, Manchester, UK
| | - Marie-Claude Asselin
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, University of Manchester, Manchester, UK
| | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Laura Michelle Parkes
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, Manchester, UK
| | - Stuart Allan
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, Manchester, UK
| | - Ingo Schiessl
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, Manchester, UK
| | - Herve Boutin
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, Manchester, UK.
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK.
| | - Ben Robert Dickie
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
20
|
Tedeschi Dauar M, Pascoal TA, Therriault J, Rowley J, Mohaddes S, Shin M, Zimmer ER, Eskildsen SF, Fonov VS, Gauthier S, Poirier J, Rosa-Neto P. Dynamic Amyloid and Metabolic Signatures of Delayed Recall Performance within the Clinical Spectrum of Alzheimer's Disease. Brain Sci 2023; 13:brainsci13020232. [PMID: 36831775 PMCID: PMC9954101 DOI: 10.3390/brainsci13020232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/31/2023] Open
Abstract
Associations between pathophysiological events and cognitive measures provide insights regarding brain networks affected during the clinical progression of Alzheimer's disease (AD). In this study, we assessed patients' scores in two delayed episodic memory tests, and investigated their associations with regional amyloid deposition and brain metabolism across the clinical spectrum of AD. We assessed the clinical, neuropsychological, structural, and positron emission tomography (PET) baseline measures of participants from the Alzheimer's Disease Neuroimaging Initiative. Subjects were classified as cognitively normal (CN), or with early (EMCI) or late (LMCI) mild cognitive impairment, or AD dementia. The memory outcome measures of interest were logical memory 30 min delayed recall (LM30) and Rey Auditory Verbal Learning Test 30 min delayed recall (RAVLT30). Voxel-based [18F]florbetapir and [18F]FDG uptake-ratio maps were constructed and correlations between PET images and cognitive scores were calculated. We found that EMCI individuals had LM30 scores negatively correlated with [18F]florbetapir uptake on the right parieto-occipital region. LMCI individuals had LM30 scores positively associated with left lateral temporal lobe [18F]FDG uptake, and RAVLT30 scores positively associated with [18F]FDG uptake in the left parietal lobe and in the right enthorhinal cortex. Additionally, LMCI individuals had LM30 scores negatively correlated with [18F]florbetapir uptake in the right frontal lobe. For the AD group, [18F]FDG uptake was positively correlated with LM30 in the left temporal lobe and with RAVLT30 in the right frontal lobe, and [18F]florbetapir uptake was negatively correlated with LM30 scores in the right parietal and left frontal lobes. The results show that the association between regional brain metabolism and the severity of episodic memory deficits is dependent on the clinical disease stage, suggesting a dynamic relationship between verbal episodic memory deficits, AD pathophysiology, and clinical disease stages.
Collapse
Affiliation(s)
- Marina Tedeschi Dauar
- Douglas Mental Health University Institute, 6875 Lasalle Boulevard, Montreal, QC H4H 1R3, Canada
- Centre for the Studies in the Prevention of Alzheimer’s Disease, 6875 Lasalle Boulevard, Montreal, QC H4H 1R3, Canada
- CAPES Foundation, Ministry of Education of Brazil, Brasilia 70040-020, DF, Brazil
- McGill University, Montreal, QC H3A 0G4, Canada
| | - Tharick Ali Pascoal
- McGill University, Montreal, QC H3A 0G4, Canada
- Translational Neuroimaging Laboratory, 6825 Lasalle Boulevard, Montreal, QC H4H 1R3, Canada
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, 3811 O’Hara St, Pittsburgh, PA 15213, USA
| | - Joseph Therriault
- McGill University, Montreal, QC H3A 0G4, Canada
- Translational Neuroimaging Laboratory, 6825 Lasalle Boulevard, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, 3801 University st., Montreal, QC H3A 2B4, Canada
| | - Jared Rowley
- Translational Neuroimaging Laboratory, 6825 Lasalle Boulevard, Montreal, QC H4H 1R3, Canada
| | - Sara Mohaddes
- Translational Neuroimaging Laboratory, 6825 Lasalle Boulevard, Montreal, QC H4H 1R3, Canada
| | - Monica Shin
- Translational Neuroimaging Laboratory, 6825 Lasalle Boulevard, Montreal, QC H4H 1R3, Canada
| | - Eduardo R. Zimmer
- Translational Neuroimaging Laboratory, 6825 Lasalle Boulevard, Montreal, QC H4H 1R3, Canada
- Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
| | - Simon Fristed Eskildsen
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Universitetsbyen 3, 8000 Aarhus, Denmark
| | - Vladimir S. Fonov
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC H3A 2B4, Canada
| | - Serge Gauthier
- McGill University, Montreal, QC H3A 0G4, Canada
- Translational Neuroimaging Laboratory, 6825 Lasalle Boulevard, Montreal, QC H4H 1R3, Canada
| | - Judes Poirier
- Douglas Mental Health University Institute, 6875 Lasalle Boulevard, Montreal, QC H4H 1R3, Canada
- Centre for the Studies in the Prevention of Alzheimer’s Disease, 6875 Lasalle Boulevard, Montreal, QC H4H 1R3, Canada
- McGill University, Montreal, QC H3A 0G4, Canada
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
| | - Pedro Rosa-Neto
- Douglas Mental Health University Institute, 6875 Lasalle Boulevard, Montreal, QC H4H 1R3, Canada
- McGill University, Montreal, QC H3A 0G4, Canada
- Translational Neuroimaging Laboratory, 6825 Lasalle Boulevard, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, 3801 University st., Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 0G4, Canada
- The McGill University Research Centre for Studies in Aging 6825 La Salle Boulevard, Montreal, QC H4H 1R3, Canada
- Correspondence:
| |
Collapse
|
21
|
Yabuuchi K, Kimura N, Masuda T, Matsubara E. Comparison of Brain Amyloid Deposition and Cortical Glucose Metabolism Between Clinic- and Community-Based Cohort. J Alzheimers Dis 2023; 95:299-306. [PMID: 37483008 DOI: 10.3233/jad-230550] [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: 07/25/2023]
Abstract
BACKGROUND The differences in positron emission tomography (PET) imaging among older adults with mild cognitive impairment (MCI), according to the recruitment source, remain unclear. OBJECTIVE To investigate the differences in brain amyloid deposition and cortical glucose metabolism according to recruitment source among older adults with MCI. METHODS Participants in the clinic-based MCI cohort, who were referred to Oita University Hospital for cognitive decline, consisted of 90 adults with MCI. The community-based MCI cohort, which participated in a prospective cohort study, consisted of 118 adults with MCI. Participants underwent cognitive function evaluation, 11C-Pittsburgh compound B (PiB)-PET, and 18F-fluorodeoxyglucose (FDG)-PET. The prevalence of amyloid positivity and mean PiB and FDG uptake values were compared between the cohorts. Moreover, a voxel-by-voxel group study was performed to determine the areas with significant differences between the clinic- and community-based MCI cohorts. RESULTS The prevalence of amyloid positivity and mean PiB uptake value in the clinic-based MCI cohort were significantly higher than those in the community-based MCI cohort (p < 0.001 and p < 0.001, respectively). The mean FDG uptake value in the clinic-based MCI cohort was significantly lower than that in the community-based MCI cohort (p < 0.001). SPM 8 analysis showed significantly increased PiB uptake in the precuneus and parietotemporal lobe and significantly decreased FDG uptake in the posterior cingulate in the clinic-based MCI cohort compared to the community-based MCI cohort. CONCLUSION The prevalence and severity of amyloid pathology in older adults with MCI varied depending on the recruitment source.
Collapse
Affiliation(s)
- Kenichi Yabuuchi
- Department of Neurology, Oita University, Faculty of Medicine, Yufu, Oita, Japan
| | - Noriyuki Kimura
- Department of Neurology, Oita University, Faculty of Medicine, Yufu, Oita, Japan
| | - Teruaki Masuda
- Department of Neurology, Oita University, Faculty of Medicine, Yufu, Oita, Japan
| | - Etsuro Matsubara
- Department of Neurology, Oita University, Faculty of Medicine, Yufu, Oita, Japan
| |
Collapse
|
22
|
Paramanick D, Singh VD, Singh VK. Neuroprotective effect of phytoconstituents via nanotechnology for treatment of Alzheimer diseases. J Control Release 2022; 351:638-655. [DOI: 10.1016/j.jconrel.2022.09.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/25/2022] [Accepted: 09/27/2022] [Indexed: 11/26/2022]
|
23
|
Chia K, Klingseisen A, Sieger D, Priller J. Zebrafish as a model organism for neurodegenerative disease. Front Mol Neurosci 2022; 15:940484. [PMID: 36311026 PMCID: PMC9606821 DOI: 10.3389/fnmol.2022.940484] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/01/2022] [Indexed: 11/20/2022] Open
Abstract
The zebrafish is increasingly recognized as a model organism for translational research into human neuropathology. The zebrafish brain exhibits fundamental resemblance with human neuroanatomical and neurochemical pathways, and hallmarks of human brain pathology such as protein aggregation, neuronal degeneration and activation of glial cells, for example, can be modeled and recapitulated in the fish central nervous system. Genetic manipulation, imaging, and drug screening are areas where zebrafish excel with the ease of introducing mutations and transgenes, the expression of fluorescent markers that can be detected in vivo in the transparent larval stages overtime, and simple treatment of large numbers of fish larvae at once followed by automated screening and imaging. In this review, we summarize how zebrafish have successfully been employed to model human neurodegenerative diseases such as Parkinson’s disease, Alzheimer’s disease, amyotrophic lateral sclerosis, and Huntington’s disease. We discuss advantages and disadvantages of choosing zebrafish as a model for these neurodegenerative conditions.
Collapse
Affiliation(s)
- Kelda Chia
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- United Kingdom Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
| | - Anna Klingseisen
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- United Kingdom Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
| | - Dirk Sieger
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- *Correspondence: Dirk Sieger,
| | - Josef Priller
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- United Kingdom Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany
- Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité - Universitätsmedizin Berlin, DZNE, Berlin, Germany
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Josef Priller,
| |
Collapse
|
24
|
Greene AN, Solomon MB, Privette Vinnedge LM. Novel molecular mechanisms in Alzheimer's disease: The potential role of DEK in disease pathogenesis. Front Aging Neurosci 2022; 14:1018180. [PMID: 36275000 PMCID: PMC9582447 DOI: 10.3389/fnagi.2022.1018180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease and age-related dementias (AD/ADRD) are debilitating diseases that exact a significant physical, emotional, cognitive, and financial toll on the individual and their social network. While genetic risk factors for early-onset AD have been identified, the molecular and genetic drivers of late-onset AD, the most common subtype, remain a mystery. Current treatment options are limited for the 35 million people in the United States with AD/ADRD. Thus, it is critically important to identify novel molecular mechanisms of dementia-related pathology that may be targets for the development of new interventions. Here, we summarize the overarching concepts regarding AD/ADRD pathogenesis. Then, we highlight one potential molecular driver of AD/ADRD, the chromatin remodeling protein DEK. We discuss in vitro, in vivo, and ex vivo findings, from our group and others, that link DEK loss with the cellular, molecular, and behavioral signatures of AD/ADRD. These include associations between DEK loss and cellular and molecular hallmarks of AD/ADRD, including apoptosis, Tau expression, and Tau hyperphosphorylation. We also briefly discuss work that suggests sex-specific differences in the role of DEK in AD/ADRD pathogenesis. Finally, we discuss future directions for exploiting the DEK protein as a novel player and potential therapeutic target for the treatment of AD/ADRD.
Collapse
Affiliation(s)
- Allie N. Greene
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Matia B. Solomon
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Department of Psychology, University of Cincinnati, Cincinnati, OH, United States
| | - Lisa M. Privette Vinnedge
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
25
|
Hvingelby VS, Højholt Terkelsen M, Johnsen EL, Møller M, Danielsen EH, Henriksen T, Glud AN, Tai Y, Møller Andersen AS, Meier K, Borghammer P, Moro E, Sørensen JCH, Pavese N. Spinal cord stimulation therapy for patients with Parkinson's disease and gait problems (STEP-PD): study protocol for an exploratory, double-blind, randomised, placebo-controlled feasibility trial. BMJ Neurol Open 2022; 4:e000333. [PMID: 36101543 PMCID: PMC9413283 DOI: 10.1136/bmjno-2022-000333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/20/2022] [Indexed: 11/04/2022] Open
Abstract
Introduction Gait difficulties are common in Parkinson's disease (PD) and cause significant disability. These symptoms are often resistant to treatment. Spinal cord stimulation (SCS) has been found to improve gait, including freezing of gait, in a small number of patients with PD. The mechanism of action is unclear, and some patients are non-responders. With this double-blind, placebo-controlled efficacy and feasibility clinical and imaging study, we aim to shed light on the mechanism of action of SCS and collect data to inform development of a scientifically sound clinical trial protocol. We also aim to identify clinical and imaging biomarkers at baseline that could be predictive of a favourable or a negative outcome of SCS and improve patient selection. Methods and analysis A total of 14 patients will be assessed with clinical rating scales and gait evaluations at baseline, and at 6 and 12 months after SCS implantation. They will also receive serial 18F-deoxyglucose and 18FEOBV PET scans to assess the effects of SCS on cortical/subcortical activity and brain cholinergic function. The first two patients will be included in an open pilot study while the rest will be randomised to receive active treatment or placebo (no stimulation) for 6 months. From this point, the entire cohort will enter an open label active treatment phase for a subsequent 6 months. Ethics and dissemination This study was reviewed and approved by the Committee on Health Research Ethics, Central Denmark RM. It is funded by the Danish Council for Independent Research. Independent of outcome, the results will be published in peer-reviewed journals and presented at national and international conferences. Trial registration number NCT05110053; ClinicalTrials.gov Identifier.
Collapse
Affiliation(s)
- Victor S Hvingelby
- Department of Clinical Medicine—Nuclear Medicine and PET Center, Aarhus University, Aarhus, Denmark
| | - Miriam Højholt Terkelsen
- Department of Clinical Medicine—Nuclear Medicine and PET Center, Aarhus University, Aarhus, Denmark
| | - Erik L Johnsen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark,Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Mette Møller
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Tove Henriksen
- Department of Neurology, Bispebjerg Hospital, Copenhagen, Denmark
| | - Andreas Nørgaard Glud
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark,Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
| | - Yen Tai
- Department of Neurosciences, Imperial College Healthcare NHS Trust, London, UK
| | | | - Kaare Meier
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark,Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
| | - Per Borghammer
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark,Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Elena Moro
- Department of Psychiatry, Neurology, Neurological Rehabilitation and Forensic Medicine, Grenoble Alpes University Hospital, Grenoble, Auvergne-Rhône-Alpes, France
| | - Jens Christian Hedemann Sørensen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark,Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
| | - Nicola Pavese
- Department of Clinical Medicine—Nuclear Medicine and PET Center, Aarhus University, Aarhus, Denmark
| |
Collapse
|
26
|
Abstract
Alzheimer’s disease (AD) is the most common major neurocognitive disorder of ageing. Although largely ignored until about a decade ago, accumulating evidence suggests that deteriorating brain energy metabolism plays a key role in the development and/or progression of AD-associated cognitive decline. Brain glucose hypometabolism is a well-established biomarker in AD but was mostly assumed to be a consequence of neuronal dysfunction and death. However, its presence in cognitively asymptomatic populations at higher risk of AD strongly suggests that it is actually a pre-symptomatic component in the development of AD. The question then arises as to whether progressive AD-related cognitive decline could be prevented or slowed down by correcting or bypassing this progressive ‘brain energy gap’. In this review, we provide an overview of research on brain glucose and ketone metabolism in AD and its prodromal condition – mild cognitive impairment (MCI) – to provide a clearer basis for proposing keto-therapeutics as a strategy for brain energy rescue in AD. We also discuss studies using ketogenic interventions and their impact on plasma ketone levels, brain energetics and cognitive performance in MCI and AD. Given that exercise has several overlapping metabolic effects with ketones, we propose that in combination these two approaches might be synergistic for brain health during ageing. As cause-and-effect relationships between the different hallmarks of AD are emerging, further research efforts should focus on optimising the efficacy, acceptability and accessibility of keto-therapeutics in AD and populations at risk of AD.
Collapse
|
27
|
Faldu KG, Shah JS. Alzheimer's disease: a scoping review of biomarker research and development for effective disease diagnosis. Expert Rev Mol Diagn 2022; 22:681-703. [PMID: 35855631 DOI: 10.1080/14737159.2022.2104639] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is regarded as the foremost reason for neurodegeneration that prominently affects the geriatric population. Characterized by extracellular accumulation of amyloid-beta (Aβ), intracellular aggregation of hyperphosphorylated tau (p-tau), and neuronal degeneration that causes impairment of memory and cognition. Amyloid/tau/neurodegeneration (ATN) classification is utilized for research purposes and involves amyloid, tau, and neuronal injury staging through MRI, PET scanning, and CSF protein concentration estimations. CSF sampling is invasive, and MRI and PET scanning requires sophisticated radiological facilities which limit its widespread diagnostic use. ATN classification lacks effectiveness in preclinical AD. AREAS COVERED This publication intends to collate and review the existing biomarker profile and the current research and development of a new arsenal of biomarkers for AD pathology from different biological samples, microRNA (miRNA), proteomics, metabolomics, artificial intelligence, and machine learning for AD screening, diagnosis, prognosis, and monitoring of AD treatments. EXPERT OPINION It is an accepted observation that AD-related pathological changes occur over a long period of time before the first symptoms are observed providing ample opportunity for detection of biological alterations in various biological samples that can aid in early diagnosis and modify treatment outcomes.
Collapse
Affiliation(s)
- Khushboo Govind Faldu
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Jigna Samir Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| |
Collapse
|
28
|
Miotto EC, Brucki SMD, Cerqueira CT, Bazán PR, Silva GADA, Martin MDGM, da Silveira PS, Faria DDP, Coutinho AM, Buchpiguel CA, Busatto Filho G, Nitrini R. Episodic Memory, Hippocampal Volume, and Function for Classification of Mild Cognitive Impairment Patients Regarding Amyloid Pathology. J Alzheimers Dis 2022; 89:181-192. [PMID: 35871330 PMCID: PMC9484090 DOI: 10.3233/jad-220100] [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] [Indexed: 11/15/2022]
Abstract
Background: Previous studies of hippocampal function and volume related to episodic memory deficits in patients with amnestic mild cognitive impairment (aMCI) have produced mixed results including increased or decreased activity and volume. However, most of them have not included biomarkers, such as amyloid-β (Aβ) deposition which is the hallmark for early identification of the Alzheimer’s disease continuum. Objective: We investigated the role of Aβ deposition, functional hippocampal activity and structural volume in aMCI patients and healthy elderly controls (HC) using a new functional MRI (fMRI) ecological episodic memory task. Methods: Forty-six older adults were included, among them Aβ PET PIB positive (PIB+) aMCI (N = 17), Aβ PET PIB negative (PIB–) aMCI (N = 15), and HC (N = 14). Hippocampal volume and function were analyzed using Freesurfer v6.0 and FSL for news headlines episodic memory fMRI task, and logistic regression for group classification in conjunction with episodic memory task and traditional neuropsychological tests. Results: The aMCI PIB+ and PIB–patients showed significantly worse performance in relation to HC in most traditional neuropsychological tests and within group difference only on story recall and the ecological episodic memory fMRI task delayed recall. The classification model reached a significant accuracy (78%) and the classification pattern characterizing the PIB+ included decreased left hippocampal function and volume, increased right hippocampal function and volume, and worse episodic memory performance differing from PIB–which showed increased left hippocampus volume. Conclusion: The main findings showed differential neural correlates, hippocampal volume and function during episodic memory in aMCI patients with the presence of Aβ deposition.
Collapse
Affiliation(s)
- Eliane Correa Miotto
- Department of Neurology, University of São Paulo, São Paulo, Brazil.,Institute of Radiology, LIM-44, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | | | | | - Paulo R Bazán
- Institute of Radiology, LIM-44, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil.,Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | - Maria da Graça M Martin
- Institute of Radiology, LIM-44, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | | | - Daniele de Paula Faria
- Laboratory of Nuclear Medicine, LIM 43, Department of Radiology and Oncology, University of Sao Paulo, Brazil
| | - Artur Martins Coutinho
- Laboratory of Nuclear Medicine, LIM 43, Department of Radiology and Oncology, University of Sao Paulo, Brazil
| | - Carlos Alberto Buchpiguel
- Laboratory of Nuclear Medicine, LIM 43, Department of Radiology and Oncology, University of Sao Paulo, Brazil
| | | | - Ricardo Nitrini
- Department of Neurology, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
29
|
Townsend RF, Woodside JV, Prinelli F, O'Neill RF, McEvoy CT. Associations Between Dietary Patterns and Neuroimaging Markers: A Systematic Review. Front Nutr 2022; 9:806006. [PMID: 35571887 PMCID: PMC9097077 DOI: 10.3389/fnut.2022.806006] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 03/16/2022] [Indexed: 12/13/2022] Open
Abstract
Dementia is a complex, growing challenge for population health worldwide. Dietary patterns (DPs) may offer an opportunity to beneficially influence cognitive ageing and potentially reduce an individuals’ risk of dementia through diet-related mechanisms. However, previous studies within this area have shown mixed results, which may be partly explained by the lack of sensitivity and accuracy within cognitive testing methods. Novel neuroimaging techniques provide a sensitive method to analyse brain changes preceding cognitive impairment which may have previously remained undetected. The purpose of this systematic review was to elucidate the role of DPs in relation to brain ageing processes, by summarising current prospective and intervention studies. Nine prospective studies met the inclusion criteria for the review, seven evaluated the Mediterranean diet (MeDi), one evaluated the Alternative Healthy Eating Index-2010, and one evaluated a posteriori derived DPs. No intervention studies were eligible for inclusion in this review. There was some evidence of an association between healthy DPs and neuroimaging markers including changes within these markers over time. Consequently, it is plausible that better adherence to such DPs may positively influence brain ageing and neurodegeneration. Future studies may benefit from the use of multi-modal neuroimaging techniques, to further investigate how adherence to a DP influences brain health. The review also highlights the crucial need for further intervention studies within this research area.
Collapse
Affiliation(s)
- Rebecca F Townsend
- Centre for Public Health, Queen's University Belfast, Belfast, United Kingdom
| | - Jayne V Woodside
- Centre for Public Health, Queen's University Belfast, Belfast, United Kingdom.,Institute for Global Food Security, Queen's University Belfast, Belfast, United Kingdom
| | - Federica Prinelli
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
| | - Roisin F O'Neill
- Centre for Public Health, Queen's University Belfast, Belfast, United Kingdom
| | - Claire T McEvoy
- Centre for Public Health, Queen's University Belfast, Belfast, United Kingdom.,Institute for Global Food Security, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
30
|
Benavides GA, Mueller T, Darley-Usmar V, Zhang J. Optimization of measurement of mitochondrial electron transport activity in postmortem human brain samples and measurement of susceptibility to rotenone and 4-hydroxynonenal inhibition. Redox Biol 2022; 50:102241. [PMID: 35066289 PMCID: PMC8792425 DOI: 10.1016/j.redox.2022.102241] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 11/25/2022] Open
Abstract
Mitochondrial function is required to meet the energetic and metabolic requirements of the brain. Abnormalities in mitochondrial function, due to genetic or developmental factors, mitochondrial toxins, aging or insufficient mitochondrial quality control contribute to neurological and psychiatric diseases. Studying bioenergetics from postmortem human tissues has been challenging due to the diverse range of human genetics, health conditions, sex, age, and postmortem interval. Furthermore, fresh tissues that were in the past required for assessment of mitochondrial respiratory function were rarely available. Recent studies established protocols to use in bioenergetic analyses from frozen tissues using animal models and cell cultures. In this study we optimized these methods to determine the activities of mitochondrial electron transport in postmortem human brain. Further we demonstrate how these samples can be used to assess the susceptibility to the mitochondrial toxin rotenone and exposure to the reactive lipid species 4-hydroxynonenal. The establishment of such an approach will significantly impact translational studies of human diseases by allowing measurement of mitochondrial function in human tissue repositories.
Collapse
Affiliation(s)
- Gloria A Benavides
- Department of Pathology, Mitochondrial Medicine Laboratory, Birmingham, AL, 35294, USA
| | - Toni Mueller
- Department of Pathology, Mitochondrial Medicine Laboratory, Birmingham, AL, 35294, USA
| | - Victor Darley-Usmar
- Department of Pathology, Mitochondrial Medicine Laboratory, Birmingham, AL, 35294, USA
| | - Jianhua Zhang
- Department of Pathology, Mitochondrial Medicine Laboratory, Birmingham, AL, 35294, USA; Birmingham VA Medical Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
31
|
McKay NS, Dincer A, Mehrotra V, Aschenbrenner AJ, Balota D, Hornbeck RC, Hassenstab J, Morris JC, Benzinger TLS, Gordon BA. Beta-amyloid moderates the relationship between cortical thickness and attentional control in middle- and older-aged adults. Neurobiol Aging 2022; 112:181-190. [PMID: 35227946 PMCID: PMC9208719 DOI: 10.1016/j.neurobiolaging.2021.12.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 12/13/2021] [Accepted: 12/28/2021] [Indexed: 12/13/2022]
Abstract
Although often unmeasured in studies of cognition, many older adults possess Alzheimer disease (AD) pathologies such as beta-amyloid (Aβ) deposition, despite being asymptomatic. We were interested in examining whether the behavior-structure relationship observed in later life was altered by the presence of preclinical AD pathology. A total of 511 cognitively unimpaired adults completed magnetic resonance imaging and three attentional control tasks; a subset (n = 396) also underwent Aβ-positron emissions tomography. A vertex-wise model was conducted to spatially represent the relationship between cortical thickness and average attentional control accuracy, while moderation analysis examined whether Aβ deposition impacted this relationship. First, we found that reduced cortical thickness in temporal, medial- and lateral-parietal, and dorsolateral prefrontal cortex, predicted worse performance on the attention task composite. Subsequent moderation analyses observed that levels of Aβ significantly influence the relationship between cortical thickness and attentional control. Our results support the hypothesis that preclinical AD, as measured by Aβ deposition, is partially driving what would otherwise be considered general aging in a cognitively normal adult population.
Collapse
Affiliation(s)
- Nicole S McKay
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; Knight Alzheimer's Disease Research Center, Washington University in St. Louis, MO.
| | - Aylin Dincer
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; Knight Alzheimer's Disease Research Center, Washington University in St. Louis, MO
| | | | - Andrew J Aschenbrenner
- Knight Alzheimer's Disease Research Center, Washington University in St. Louis, MO; Department of Neurology, Washington School of Medicine, St. Louis, MO
| | - David Balota
- Knight Alzheimer's Disease Research Center, Washington University in St. Louis, MO; Department of Psychological and Brain Sciences, Washington University in St. Louis, MO
| | - Russ C Hornbeck
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; Knight Alzheimer's Disease Research Center, Washington University in St. Louis, MO
| | - Jason Hassenstab
- Knight Alzheimer's Disease Research Center, Washington University in St. Louis, MO; Department of Neurology, Washington School of Medicine, St. Louis, MO; Department of Psychological and Brain Sciences, Washington University in St. Louis, MO
| | - John C Morris
- Knight Alzheimer's Disease Research Center, Washington University in St. Louis, MO; Department of Neurology, Washington School of Medicine, St. Louis, MO
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; Knight Alzheimer's Disease Research Center, Washington University in St. Louis, MO
| | - Brian A Gordon
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; Knight Alzheimer's Disease Research Center, Washington University in St. Louis, MO; Department of Psychological and Brain Sciences, Washington University in St. Louis, MO
| |
Collapse
|
32
|
Soloperto A, Quaglio D, Baiocco P, Romeo I, Mori M, Ardini M, Presutti C, Sannino I, Ghirga S, Iazzetti A, Ippoliti R, Ruocco G, Botta B, Ghirga F, Di Angelantonio S, Boffi A. Rational design and synthesis of a novel BODIPY-based probe for selective imaging of tau tangles in human iPSC-derived cortical neurons. Sci Rep 2022; 12:5257. [PMID: 35347170 PMCID: PMC8960764 DOI: 10.1038/s41598-022-09016-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 03/15/2022] [Indexed: 12/26/2022] Open
Abstract
Numerous studies have shown a strong correlation between the number of neurofibrillary tangles of the tau protein and Alzheimer's disease progression, making the quantitative detection of tau very promising from a clinical point of view. However, the lack of highly reliable fluorescent probes for selective imaging of tau neurofibrillary tangles is a major challenge due to sharing similar β–sheet motifs with homologous Amyloid-β fibrils. In the current work, we describe the rational design and the in silico evaluation of a small-size focused library of fluorescent probes, consisting of a BODIPY core (electron acceptor) featuring highly conjugated systems (electron donor) with a length in the range 13–19 Å at C3. Among the most promising probes in terms of binding mode, theoretical affinity and polarity, BT1 has been synthesized and tested in vitro onto human induced pluripotent stem cells derived neuronal cell cultures. The probe showed excellent photophysical properties and high selectivity allowing in vitro imaging of hyperphosphorylated tau protein filaments with minimal background noise. Our findings offer new insight into the structure-activity relationship of this class of tau selective fluorophores, paving the way for boosting tau tangle detection in patients possibly through retinal spectral scans.
Collapse
Affiliation(s)
- Alessandro Soloperto
- Center for Life Nano- & Neuro-Science, Istituto Italiano Di Tecnologia, 00161, Rome, Italy
| | - Deborah Quaglio
- Department of Chemistry and Technology of Drugs, Department of Excellence 2018-2022, Sapienza University of Rome, 00185, Rome, Italy
| | - Paola Baiocco
- Center for Life Nano- & Neuro-Science, Istituto Italiano Di Tecnologia, 00161, Rome, Italy.,Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, 00185, Rome, Italy
| | - Isabella Romeo
- Center for Life Nano- & Neuro-Science, Istituto Italiano Di Tecnologia, 00161, Rome, Italy.,Department of Chemistry and Technology of Drugs, Department of Excellence 2018-2022, Sapienza University of Rome, 00185, Rome, Italy
| | - Mattia Mori
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, 53100, Siena, Italy
| | - Matteo Ardini
- Department of Life, Health, and Environmental Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Caterina Presutti
- Center for Life Nano- & Neuro-Science, Istituto Italiano Di Tecnologia, 00161, Rome, Italy.,Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, 00185, Rome, Italy
| | - Ida Sannino
- Center for Life Nano- & Neuro-Science, Istituto Italiano Di Tecnologia, 00161, Rome, Italy
| | - Silvia Ghirga
- Center for Life Nano- & Neuro-Science, Istituto Italiano Di Tecnologia, 00161, Rome, Italy
| | - Antonia Iazzetti
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of Sacred Heart, 00168, Rome, Italy
| | - Rodolfo Ippoliti
- Department of Life, Health, and Environmental Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Giancarlo Ruocco
- Center for Life Nano- & Neuro-Science, Istituto Italiano Di Tecnologia, 00161, Rome, Italy
| | - Bruno Botta
- Department of Chemistry and Technology of Drugs, Department of Excellence 2018-2022, Sapienza University of Rome, 00185, Rome, Italy
| | - Francesca Ghirga
- Department of Chemistry and Technology of Drugs, Department of Excellence 2018-2022, Sapienza University of Rome, 00185, Rome, Italy.
| | - Silvia Di Angelantonio
- Center for Life Nano- & Neuro-Science, Istituto Italiano Di Tecnologia, 00161, Rome, Italy. .,Department of Physiology and Pharmacology, Sapienza University of Rome, 00185, Rome, Italy.
| | - Alberto Boffi
- Center for Life Nano- & Neuro-Science, Istituto Italiano Di Tecnologia, 00161, Rome, Italy.,Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, 00185, Rome, Italy
| |
Collapse
|
33
|
An efficient combination of quadruple biomarkers in binary classification using ensemble machine learning technique for early onset of Alzheimer disease. Neural Comput Appl 2022. [DOI: 10.1007/s00521-022-07076-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
34
|
Dodson M, Benavides GA, Darley-Usmar V, Zhang J. Differential Effects of 2-Deoxyglucose and Glucose Deprivation on 4-Hydroxynonenal Dependent Mitochondrial Dysfunction in Primary Neurons. FRONTIERS IN AGING 2022; 3:812810. [PMID: 35821809 PMCID: PMC9261388 DOI: 10.3389/fragi.2022.812810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/26/2022] [Indexed: 11/13/2022]
Abstract
Mitochondrial dysfunction and metabolic decline are prevalent features of aging and age-related disorders, including neurodegeneration. Neurodegenerative diseases are associated with a progressive loss of metabolic homeostasis. This pathogenic decline in metabolism is the result of several factors, including decreased mitochondrial function, increased oxidative stress, inhibited autophagic flux, and altered metabolic substrate availability. One critical metabolite for maintaining neuronal function is glucose, which is utilized by the brain more than any other organ to meet its substantial metabolic demand. Enzymatic conversion of glucose into its downstream metabolites is critical for maintaining neuronal cell growth and overall metabolic homeostasis. Perturbation of glycolysis could significantly hinder neuronal metabolism by affecting key metabolic pathways. Here, we demonstrate that the glucose analogue 2-deoxyglucose (2DG) decreases cell viability, as well as both basal and maximal mitochondrial oxygen consumption in response to the neurotoxic lipid 4-hydroxynonenal (HNE), whereas glucose deprivation has a minimal effect. Furthermore, using a cell permeabilization assay we found that 2DG has a more pronounced effect on HNE-dependent inhibition of mitochondrial complex I and II than glucose deprivation. Importantly, these findings indicate that altered glucose utilization plays a critical role in dictating neuronal survival by regulating the mitochondrial response to electrophilic stress.
Collapse
Affiliation(s)
- Matthew Dodson
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Gloria A. Benavides
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Victor Darley-Usmar
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jianhua Zhang
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Veterans Affairs, Birmingham VA Medical Center, University of Alabama at Birmingham, Birmingham, AL, United States
- *Correspondence: Jianhua Zhang,
| |
Collapse
|
35
|
Jeong H, Song IU, Chung YA, Park JS, Na SH, Im JJ, Bikson M, Lee W, Yoo SS. Short-Term Efficacy of Transcranial Focused Ultrasound to the Hippocampus in Alzheimer’s Disease: A Preliminary Study. J Pers Med 2022; 12:jpm12020250. [PMID: 35207738 PMCID: PMC8878180 DOI: 10.3390/jpm12020250] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/27/2022] [Accepted: 02/03/2022] [Indexed: 11/17/2022] Open
Abstract
Preclinical studies have suggested that low-intensity transcranial focused ultrasound (tFUS) may have therapeutic potential for Alzheimer’s disease (AD) by opening the blood–brain barrier (BBB), reducing amyloid pathology, and improving cognition. This study investigated the effects of tFUS on BBB opening, regional cerebral metabolic rate of glucose (rCMRglu), and cognitive function in AD patients. Eight patients with AD received image-guided tFUS to the right hippocampus immediately after intravenous injection of microbubble ultrasound contrast agents. Patients completed magnetic resonance imaging (MRI), 18F-fluoro-2-deoxyglucose positron emission tomography (PET), and cognitive assessments before and after the sonication. No evidence of transient BBB opening was found on T1 dynamic contrast-enhanced MRI. However, immediate recall (p = 0.03) and recognition memory (p = 0.02) were significantly improved on the verbal learning test. PET image analysis demonstrated increased rCMRglu in the right hippocampus (p = 0.001). In addition, increases of hippocampal rCMRglu were correlated with improvement in recognition memory (Spearman’s ρ = 0.77, p = 0.02). No adverse event was observed. Our results suggest that tFUS to the hippocampus of AD patients may improve rCMRglu of the target area and memory in the short term, even without BBB opening. Further larger sham-controlled trials with loger follow-up are warranted to evaluate the efficacy and safety of tFUS in patients with AD.
Collapse
Affiliation(s)
- Hyeonseok Jeong
- Department of Nuclear Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 21431, Korea; (H.J.); (J.J.I.)
- Department of Radiology, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 21431, Korea
| | - In-Uk Song
- Department of Neurology, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 21431, Korea; (J.-S.P.); (S.-H.N.)
- Correspondence: (I.-U.S.); (Y.-A.C.); Tel.: +82-32-280-5010 (I.-U.S.); +82-32-280-5242 (Y.-A.C.); Fax: +82-32-280-5244 (I.-U.S. & Y.-A.C.)
| | - Yong-An Chung
- Department of Nuclear Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 21431, Korea; (H.J.); (J.J.I.)
- Department of Radiology, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 21431, Korea
- Correspondence: (I.-U.S.); (Y.-A.C.); Tel.: +82-32-280-5010 (I.-U.S.); +82-32-280-5242 (Y.-A.C.); Fax: +82-32-280-5244 (I.-U.S. & Y.-A.C.)
| | - Jong-Sik Park
- Department of Neurology, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 21431, Korea; (J.-S.P.); (S.-H.N.)
| | - Seung-Hee Na
- Department of Neurology, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 21431, Korea; (J.-S.P.); (S.-H.N.)
| | - Jooyeon Jamie Im
- Department of Nuclear Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 21431, Korea; (H.J.); (J.J.I.)
| | - Marom Bikson
- Department of Biomedical Engineering, The City College of New York, New York, NY 10031, USA;
| | - Wonhye Lee
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (W.L.); (S.-S.Y.)
| | - Seung-Schik Yoo
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (W.L.); (S.-S.Y.)
| |
Collapse
|
36
|
Austad SN, Ballinger S, Buford TW, Carter CS, Smith DL, Darley-Usmar V, Zhang J. Targeting whole body metabolism and mitochondrial bioenergetics in the drug development for Alzheimer's disease. Acta Pharm Sin B 2022; 12:511-531. [PMID: 35256932 PMCID: PMC8897048 DOI: 10.1016/j.apsb.2021.06.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/26/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
Aging is by far the most prominent risk factor for Alzheimer's disease (AD), and both aging and AD are associated with apparent metabolic alterations. As developing effective therapeutic interventions to treat AD is clearly in urgent need, the impact of modulating whole-body and intracellular metabolism in preclinical models and in human patients, on disease pathogenesis, have been explored. There is also an increasing awareness of differential risk and potential targeting strategies related to biological sex, microbiome, and circadian regulation. As a major part of intracellular metabolism, mitochondrial bioenergetics, mitochondrial quality-control mechanisms, and mitochondria-linked inflammatory responses have been considered for AD therapeutic interventions. This review summarizes and highlights these efforts.
Collapse
Key Words
- ACE2, angiotensin I converting enzyme (peptidyl-dipeptidase A) 2
- AD, Alzheimer's disease
- ADP, adenosine diphosphate
- ADRD, AD-related dementias
- Aβ, amyloid β
- CSF, cerebrospinal fluid
- Circadian regulation
- DAMPs
- DAMPs, damage-associated molecular patterns
- Diabetes
- ER, estrogen receptor
- ETC, electron transport chain
- FCCP, trifluoromethoxy carbonylcyanide phenylhydrazone
- FPR-1, formyl peptide receptor 1
- GIP, glucose-dependent insulinotropic polypeptide
- GLP-1, glucagon-like peptide-1
- HBP, hexoamine biosynthesis pathway
- HTRA, high temperature requirement A
- Hexokinase biosynthesis pathway
- I3A, indole-3-carboxaldehyde
- IRF-3, interferon regulatory factor 3
- LC3, microtubule associated protein light chain 3
- LPS, lipopolysaccharide
- LRR, leucine-rich repeat
- MAVS, mitochondrial anti-viral signaling
- MCI, mild cognitive impairment
- MRI, magnetic resonance imaging
- MRS, magnetic resonance spectroscopy
- Mdivi-1, mitochondrial division inhibitor 1
- Microbiome
- Mitochondrial DNA
- Mitochondrial electron transport chain
- Mitochondrial quality control
- NLRP3, leucine-rich repeat (LRR)-containing protein (NLR)-like receptor family pyrin domain containing 3
- NOD, nucleotide-binding oligomerization domain
- NeuN, neuronal nuclear protein
- PET, fluorodeoxyglucose (FDG)-positron emission tomography
- PKA, protein kinase A
- POLβ, the base-excision repair enzyme DNA polymerase β
- ROS, reactive oxygen species
- Reactive species
- SAMP8, senescence-accelerated mice
- SCFAs, short-chain fatty acids
- SIRT3, NAD-dependent deacetylase sirtuin-3
- STING, stimulator of interferon genes
- STZ, streptozotocin
- SkQ1, plastoquinonyldecyltriphenylphosphonium
- T2D, type 2 diabetes
- TCA, Tricarboxylic acid
- TLR9, toll-like receptor 9
- TMAO, trimethylamine N-oxide
- TP, tricyclic pyrone
- TRF, time-restricted feeding
- cAMP, cyclic adenosine monophosphate
- cGAS, cyclic GMP/AMP synthase
- hAPP, human amyloid precursor protein
- hPREP, human presequence protease
- i.p., intraperitoneal
- mTOR, mechanistic target of rapamycin
- mtDNA, mitochondrial DNA
- αkG, alpha-ketoglutarate
Collapse
Affiliation(s)
- Steven N. Austad
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Scott Ballinger
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Thomas W. Buford
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Christy S. Carter
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Daniel L. Smith
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Victor Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
37
|
Lack of association between cortical amyloid deposition and glucose metabolism in early stage Alzheimer´s disease patients. Radiol Oncol 2021; 56:23-31. [PMID: 34957735 PMCID: PMC8884854 DOI: 10.2478/raon-2021-0051] [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: 10/12/2021] [Accepted: 11/09/2021] [Indexed: 11/20/2022] Open
Abstract
Background Beta amyloid (Aβ) causes synaptic dysfunction leading to neuronal death. It is still controversial if the magnitude of Aβ deposition correlates with the degree of cognitive impairment. Diagnostic imaging may lead to a better understanding the role of Aβ in development of cognitive deficits. The aim of the present study was to investigate if Aβ deposition in the corresponding brain region of early stage Alzheimer´s disease (AD) patients, directly correlates to neuronal dysfunction and cognitive impairment indicated by reduced glucose metabolism. Patients and methods In 30 patients with a clinical phenotype of AD and amyloid positive brain imaging, 2-[18F] fluoro-2-deoxy-d-glucose (FDG) PET/CT was performed. We extracted the average [18F] flutemetamol (Vizamyl) uptake for each of the 16 regions of interest in both hemispheres and computed the standardized uptake value ratio (SUVR) by dividing the Vimazyl intensities by the mean signal of positive and negative control regions. Data were analysed using the R environment for statistical computing and graphics. Results Any negative correlation between Aβ deposition and glucose metabolism in 32 dementia related and corresponding brain regions in AD patients was not found. None of the correlation coefficient values were statistically significant different from zero based on two-sided p- value. Conclusions Regional Aβ deposition did not correlate negatively with local glucose metabolism in early stage AD patients. Our findings support the role of Aβ as a valid biomarker, but does not permit to conclude that Aβ is a direct cause for an aberrant brain glucose metabolism and neuronal dysfunction.
Collapse
|
38
|
Tournissac M, Leclerc M, Valentin-Escalera J, Vandal M, Bosoi CR, Planel E, Calon F. Metabolic determinants of Alzheimer's disease: A focus on thermoregulation. Ageing Res Rev 2021; 72:101462. [PMID: 34534683 DOI: 10.1016/j.arr.2021.101462] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/09/2021] [Accepted: 09/11/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a complex age-related neurodegenerative disease, associated with central and peripheral metabolic anomalies, such as impaired glucose utilization and insulin resistance. These observations led to a considerable interest not only in lifestyle-related interventions, but also in repurposing insulin and other anti-diabetic drugs to prevent or treat dementia. Body temperature is the oldest known metabolic readout and mechanisms underlying its maintenance fail in the elderly, when the incidence of AD rises. This raises the possibility that an age-associated thermoregulatory deficit contributes to energy failure underlying AD pathogenesis. Brown adipose tissue (BAT) plays a central role in thermogenesis and maintenance of body temperature. In recent years, the modulation of BAT activity has been increasingly demonstrated to regulate energy expenditure, insulin sensitivity and glucose utilization, which could also provide benefits for AD. Here, we review the evidence linking thermoregulation, BAT and insulin-related metabolic defects with AD, and we propose mechanisms through which correcting thermoregulatory impairments could slow the progression and delay the onset of AD.
Collapse
|
39
|
Zaletel I, Milutinović K, Bajčetić M, Nowakowski RS. Differentiation of Amyloid Plaques Between Alzheimer's Disease and Non-Alzheimer's Disease Individuals Based on Gray-Level Co-occurrence Matrix Texture Analysis. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2021; 27:1146-1153. [PMID: 35105417 DOI: 10.1017/s1431927621012095] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Amyloid plaques, one of the main hallmarks of Alzheimer's disease (AD), are classified into diffuse (associated with cognitive impairment) and dense-core types (a common finding in brains of people without Alzheimer's disease (non-AD) and without impaired cognitive function) based on their morphology. We tried to determine the usability of gray-level co-occurrence matrix (GLCM) texture parameters of homogeneity and heterogeneity for the differentiation of amyloid plaque images obtained from AD and non-AD individuals. Images of amyloid-β (Aβ) immunostained brain tissue samples were obtained from the Aging, Dementia and Traumatic Brain Injury Project. A total of 1,039 plaques were isolated from different brain regions of 69 AD and non-AD individuals and used for further GLCM analysis. Images of Aβ stained plaques show higher values of heterogeneity parameters and lower values of homogeneity parameters in AD patients, and vice versa in non-AD patients. Additionally, GLCM analysis shows differences in Aβ plaque texture between different brain regions in non-AD patients and correlates with variables that characterize patient's dementia status. The present study shows that GLCM texture analysis is an efficient method to discriminate between different types of amyloid plaques based on their morphology and thus can prove as a valuable tool in the neuropathological investigation of dementia.
Collapse
Affiliation(s)
- Ivan Zaletel
- Faculty of Medicine, Institute of Histology and Embryology "Aleksandar Đ. Kostić", University of Belgrade, Belgrade11000, Republic of Serbia
| | - Katarina Milutinović
- Faculty of Medicine, Institute of Histology and Embryology "Aleksandar Đ. Kostić", University of Belgrade, Belgrade11000, Republic of Serbia
| | - Miloš Bajčetić
- Faculty of Medicine, Institute of Histology and Embryology "Aleksandar Đ. Kostić", University of Belgrade, Belgrade11000, Republic of Serbia
| | - Richard S Nowakowski
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, 32306-4300, FL, USA
| |
Collapse
|
40
|
Joseph S, Knezevic D, Zomorrodi R, Blumberger DM, Daskalakis ZJ, Mulsant BH, Pollock BG, Voineskos A, Wang W, Rajji TK, Kumar S. Dorsolateral prefrontal cortex excitability abnormalities in Alzheimer's Dementia: Findings from transcranial magnetic stimulation and electroencephalography study. Int J Psychophysiol 2021; 169:55-62. [PMID: 34499960 DOI: 10.1016/j.ijpsycho.2021.08.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/04/2021] [Accepted: 08/30/2021] [Indexed: 02/06/2023]
Abstract
There is some evidence of cortical hyper-excitability in Alzheimer's Dementia (AD) but its relationship with cognition is not clear. In this study, we assessed dorsolateral prefrontal cortex (DLPFC) excitability and its relationship with cognition in AD. Twenty-four participants with AD (mean [SD] age = 74.1 [7.2] years) and eleven elderly healthy controls (HC) (mean [SD] age = 68.8 [7.3] years) were recruited. Transcranial magnetic stimulation (TMS) combined with electroencephalography (EEG) was used to assess cortical excitability. Cortical evoked activity (CEA) between 25 and 80 ms post-TMS stimulus was calculated as the primary measure of cortical excitability. TMS-evoked potential peak (TEP) amplitudes (P30, N45 and P60) were also calculated. Cognition was assessed using Montreal Cognitive Assessment (MoCA), Executive Interview (EXIT) and Cambridge Neuropsychological Test Automated Battery Stockings of Cambridge (SOC). There was no difference in TMS stimulus intensity between the groups. DLPFC-CEA was higher in the AD (mean [SD] = 134.64 [90.22] μV) than the HC group (mean [SD] = 82.65 [40.28] μV; t33 = 2.357, p = 0.025). There were no differences in TEP peak amplitudes between the groups. Further, DLPFC-CEA was inversely associated with MoCA and SOC, and positively associated with EXIT scores in AD. These results suggest increased DLPFC excitability in AD, and its inverse associations with global cognition and executive function. Future studies should examine these findings in larger samples and longitudinally, and could also assess these markers of cortical excitability in relation to other established markers of AD and in response to interventions.
Collapse
Affiliation(s)
- Shaylyn Joseph
- Centre for Addiction and Mental Health, Toronto, Canada; Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | | | - Reza Zomorrodi
- Centre for Addiction and Mental Health, Toronto, Canada; Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Daniel M Blumberger
- Centre for Addiction and Mental Health, Toronto, Canada; Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Toronto Dementia Research Alliance, Toronto, Canada
| | | | - Benoit H Mulsant
- Centre for Addiction and Mental Health, Toronto, Canada; Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Toronto Dementia Research Alliance, Toronto, Canada
| | - Bruce G Pollock
- Centre for Addiction and Mental Health, Toronto, Canada; Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Toronto Dementia Research Alliance, Toronto, Canada
| | - Aristotle Voineskos
- Centre for Addiction and Mental Health, Toronto, Canada; Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Toronto Dementia Research Alliance, Toronto, Canada
| | - Wei Wang
- Centre for Addiction and Mental Health, Toronto, Canada; University of South Florida, FL, United States
| | - Tarek K Rajji
- Centre for Addiction and Mental Health, Toronto, Canada; Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Toronto Dementia Research Alliance, Toronto, Canada
| | - Sanjeev Kumar
- Centre for Addiction and Mental Health, Toronto, Canada; Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Toronto Dementia Research Alliance, Toronto, Canada.
| |
Collapse
|
41
|
Joseph S, Patterson R, Wang W, Blumberger DM, Rajji T, Kumar S. Quantitative Assessment of Cortical Excitability in Alzheimer's Dementia and Its Association with Clinical Symptoms: A Systematic Review and Meta-Analyses. J Alzheimers Dis 2021; 88:867-891. [PMID: 34219724 DOI: 10.3233/jad-210311] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by cognitive and neuropsychiatric symptoms (NPS) due to underlying neurodegenerative pathology. Some studies using electroencephalography (EEG) have shown increased epileptiform and epileptic activity in AD. OBJECTIVE This review and meta-analyses aims to synthesize the existing evidence for quantitative abnormalities of cortical excitability in AD and their relationship with clinical symptoms. METHODS We systematically searched and reviewed publications that quantitatively assessed cortical excitability, using transcranial magnetic stimulation (TMS) resting motor threshold (rMT), active motor threshold (aMT), motor evoked potential (MEP) or directly from the cortex using TMS-EEG via TMS-evoked potential (TEP). We meta-analyzed studies that assessed rMT and aMT using random effects model. RESULTS We identified 895 publications out of which 37 were included in the qualitative review and 30 studies using rMT or aMT were included in the meta-analyses. The AD group had reduced rMT (Hedges' g = -0.99, 95%CI [-1.29, -0.68], p < 0.00001) and aMT (Hedges' g = -0.87, 95%CI [-1.50, -0.24], p < 0.00001) as compared with control groups, indicative of higher cortical excitability. Qualitative review found some evidence of increased MEP amplitude, whereas findings related to TEP were inconsistent. There was some evidence supporting an inverse association between cortical excitability and global cognition. No publications reported on the relationship between cortical excitability and NPS. CONCLUSION There is strong evidence of increased motor cortex excitability in AD and some evidence of an inverse association between excitability and cognition. Future studies should assess cortical excitability from non-motor areas using TMS-EEG and examine its relationship with cognition and NPS.
Collapse
Affiliation(s)
- Shaylyn Joseph
- Centre for Addiction and Mental Health, Toronto, Canada.,University of Toronto, Toronto, Canada
| | - Rachel Patterson
- Centre for Addiction and Mental Health, Toronto, Canada.,University of Toronto, Toronto, Canada
| | - Wei Wang
- Centre for Addiction and Mental Health, Toronto, Canada
| | - Daniel M Blumberger
- Centre for Addiction and Mental Health, Toronto, Canada.,University of Toronto, Toronto, Canada
| | - Tarek Rajji
- Centre for Addiction and Mental Health, Toronto, Canada.,University of Toronto, Toronto, Canada.,Toronto Dementia Research Alliance, Toronto, Canada
| | - Sanjeev Kumar
- Centre for Addiction and Mental Health, Toronto, Canada.,University of Toronto, Toronto, Canada
| |
Collapse
|
42
|
Kim H, Chung JY. Pathobiolgy and Management of Alzheimer's Disease. Chonnam Med J 2021; 57:108-117. [PMID: 34123738 PMCID: PMC8167446 DOI: 10.4068/cmj.2021.57.2.108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 01/02/2023] Open
Abstract
Amyloid and tau protein abnormalities have been identified as the main causes of Alzheimer's disease but exact mechanisms remain to be revealed. Especially, amyloid beta and tau protein coupling and neuroinflammatory and neurovascular contributions to Alzheimer disease are quite mysterious. Many animal models and basic biological research are trying to solve these puzzles. Known as aging processes, autophagy, mitochondrial degeneration with generation of reactive oxygen species, and age-related epigenetic modifications are also known to be associated with development of Alzheimer's disease. Environmental factors such as bacterial and viral infections, heavy metal ions, diet, sleep, stress, and gut microbiota are also risk factors of Alzheimer's disease. Future development of preventive and therapeutic modalities may be dependent on the pathobiology of Alzheimer's disease.
Collapse
Affiliation(s)
- Hoowon Kim
- Department of Neurology, Chosun University Hospital, Gwangju, Korea
| | - Ji Yeon Chung
- Department of Neurology, Chosun University Hospital, Gwangju, Korea
| |
Collapse
|
43
|
Jung ME. A Protective Role of Translocator Protein in Alzheimer's Disease Brain. Curr Alzheimer Res 2021; 17:3-15. [PMID: 32065102 DOI: 10.2174/1567205017666200217105950] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 01/13/2020] [Accepted: 01/20/2020] [Indexed: 12/16/2022]
Abstract
Translocator Protein (18 kDa) (TSPO) is a mitochondrial protein that locates cytosol cholesterol to mitochondrial membranes to begin the synthesis of steroids including neurotrophic neurosteroids. TSPO is abundantly present in glial cells that support neurons and respond to neuroinflammation. Located at the outer membrane of mitochondria, TSPO regulates the opening of mitochondrial permeability transition pore (mPTP) that controls the entry of molecules necessary for mitochondrial function. TSPO is linked to neurodegenerative Alzheimer's Disease (AD) such that TSPO is upregulated in the brain of AD patients and signals AD-induced adverse changes in brain. The initial increase in TSPO in response to brain insults remains elevated to repair cellular damages and perhaps to prevent further neuronal degeneration as AD progresses. To exert such protective activities, TSPO increases the synthesis of neuroprotective steroids, decreases neuroinflammation, limits the opening of mPTP, and reduces the generation of reactive oxygen species. The beneficial effects of TSPO on AD brain are manifested as the attenuation of neurotoxic amyloid β and mitochondrial dysfunction accompanied by the improvement of memory and cognition. However, the protective activities of TSPO appear to be temporary and eventually diminish as the severity of AD becomes profound. Timely treatment with TSPO agonists/ligands before the loss of endogenous TSPO's activity may promote the protective functions and may extend neuronal survival.
Collapse
Affiliation(s)
- Marianna E Jung
- Pharmacology and Neuroscience, University of North Texas Health Science Center, Institute for Healthy Aging, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, United States
| |
Collapse
|
44
|
Bao W, Xie F, Zuo C, Guan Y, Huang YH. PET Neuroimaging of Alzheimer's Disease: Radiotracers and Their Utility in Clinical Research. Front Aging Neurosci 2021; 13:624330. [PMID: 34025386 PMCID: PMC8134674 DOI: 10.3389/fnagi.2021.624330] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/23/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's Disease (AD), the leading cause of senile dementia, is a progressive neurodegenerative disorder affecting millions of people worldwide and exerting tremendous socioeconomic burden on all societies. Although definitive diagnosis of AD is often made in the presence of clinical manifestations in late stages, it is now universally believed that AD is a continuum of disease commencing from the preclinical stage with typical neuropathological alterations appearing decades prior to its first symptom, to the prodromal stage with slight symptoms of amnesia (amnestic mild cognitive impairment, aMCI), and then to the terminal stage with extensive loss of basic cognitive functions, i.e., AD-dementia. Positron emission tomography (PET) radiotracers have been developed in a search to meet the increasing clinical need of early detection and treatment monitoring for AD, with reference to the pathophysiological targets in Alzheimer's brain. These include the pathological aggregations of misfolded proteins such as β-amyloid (Aβ) plagues and neurofibrillary tangles (NFTs), impaired neurotransmitter system, neuroinflammation, as well as deficient synaptic vesicles and glucose utilization. In this article we survey the various PET radiotracers available for AD imaging and discuss their clinical applications especially in terms of early detection and cognitive relevance.
Collapse
Affiliation(s)
- Weiqi Bao
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Fang Xie
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Chuantao Zuo
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Yiyun Henry Huang
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
45
|
Moscoso A, Grothe MJ, Ashton NJ, Karikari TK, Lantero Rodríguez J, Snellman A, Suárez-Calvet M, Blennow K, Zetterberg H, Schöll M. Longitudinal Associations of Blood Phosphorylated Tau181 and Neurofilament Light Chain With Neurodegeneration in Alzheimer Disease. JAMA Neurol 2021; 78:396-406. [PMID: 33427873 PMCID: PMC7802009 DOI: 10.1001/jamaneurol.2020.4986] [Citation(s) in RCA: 154] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Question What is the potential of blood-based biomarkers for predicting and monitoring the progression of Alzheimer disease neurodegeneration? Findings In this cohort study that included 1113 participants from the multicentric Alzheimer’s Disease Neuroimaging Initiative study, baseline and longitudinal increases of tau phosphorylated at threonine 181 (p-tau181) in blood plasma were associated with progressive, longitudinal neurodegeneration in brain regions characteristic for Alzheimer disease, as well as with cognitive decline, only among participants with elevated brain amyloid-β. Neurofilament light chain in plasma, however, was associated with disease progression independent of amyloid-β and plasma p-tau181. Meaning These findings suggest that plasma p-tau181, alone or combined with plasma neurofilament light chain, can be used as an accessible, minimally invasive biomarker to track Alzheimer disease progression. Importance Plasma phosphorylated tau at threonine 181 (p-tau181) has been proposed as an easily accessible biomarker for the detection of Alzheimer disease (AD) pathology, but its ability to monitor disease progression in AD remains unclear. Objective To study the potential of longitudinal plasma p-tau181 measures for assessing neurodegeneration progression and cognitive decline in AD in comparison to plasma neurofilament light chain (NfL), a disease-nonspecific marker of neuronal injury. Design, Setting, and Participants This longitudinal cohort study included data from the Alzheimer’s Disease Neuroimaging Initiative from February 1, 2007, to June 6, 2016. Follow-up blood sampling was performed for up to 8 years. Plasma p-tau181 measurements were performed in 2020. This was a multicentric observational study of 1113 participants, including cognitively unimpaired participants as well as patients with cognitive impairment (mild cognitive impairment and AD dementia). Participants were eligible for inclusion if they had available plasma p-tau181 and NfL measurements and at least 1 fluorine-18–labeled fluorodeoxyglucose (FDG) positron emission tomography (PET) or structural magnetic resonance imaging scan performed at the same study visit. Exclusion criteria included any significant neurologic disorder other than suspected AD; presence of infection, infarction, or multiple lacunes as detected by magnetic resonance imaging; and any significant systemic condition that could lead to difficulty complying with the protocol. Exposures Plasma p-tau181 and NfL measured with single-molecule array technology. Main Outcomes and Measures Longitudinal imaging markers of neurodegeneration (FDG PET and structural magnetic resonance imaging) and cognitive test scores (Preclinical Alzheimer Cognitive Composite and Alzheimer Disease Assessment Scale–Cognitive Subscale with 13 tasks). Data were analyzed from June 20 to August 15, 2020. Results Of the 1113 participants (mean [SD] age, 74.0 [7.6] years; 600 men [53.9%]; 992 non-Hispanic White participants [89.1%]), a total of 378 individuals (34.0%) were cognitively unimpaired (CU) and 735 participants (66.0%) were cognitively impaired (CImp). Of the CImp group, 537 (73.1%) had mild cognitive impairment, and 198 (26.9%) had AD dementia. Longitudinal changes of plasma p-tau181 were associated with cognitive decline (CU: r = –0.24, P < .001; CImp: r = 0.34, P < .001) and a prospective decrease in glucose metabolism (CU: r = –0.05, P = .48; CImp: r = –0.27, P < .001) and gray matter volume (CU: r = –0.19, P < .001; CImp: r = –0.31, P < .001) in highly AD-characteristic brain regions. These associations were restricted to amyloid-β–positive individuals. Both plasma p-tau181 and NfL were independently associated with cognition and neurodegeneration in brain regions typically affected in AD. However, NfL was also associated with neurodegeneration in brain regions exceeding this AD-typical spatial pattern in amyloid-β–negative participants. Mediation analyses found that approximately 25% to 45% of plasma p-tau181 outcomes on cognition measures were mediated by the neuroimaging-derived markers of neurodegeneration, suggesting links between plasma p-tau181 and cognition independent of these measures. Conclusions and Relevance Study findings suggest that plasma p-tau181 was an accessible and scalable marker for predicting and monitoring neurodegeneration and cognitive decline and was, unlike plasma NfL, AD specific. The study findings suggest implications for the use of plasma biomarkers as measures to monitor AD progression in clinical practice and treatment trials.
Collapse
Affiliation(s)
- Alexis Moscoso
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Michel J Grothe
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Unidad de Trastornos del Movimiento, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, United Kingdom
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Juan Lantero Rodríguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anniina Snellman
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Turku PET Centre, University of Turku, Turku, Finland
| | - Marc Suárez-Calvet
- Barcelonaßeta Brain Research Center, Pasqual Maragall Foundation. Barcelona, Spain.,Hospital del Mar Medical Research Institute, Barcelona, Spain.,Servei de Neurologia, Hospital del Mar, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable, Madrid, Spain
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,UK Dementia Research Institute at University College London, London, United Kingdom
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | | |
Collapse
|
46
|
Femminella GD, Livingston NR, Raza S, van der Doef T, Frangou E, Love S, Busza G, Calsolaro V, Carver S, Holmes C, Ritchie CW, Lawrence RM, McFarlane B, Tadros G, Ridha BH, Bannister C, Walker Z, Archer H, Coulthard E, Underwood B, Prasanna A, Koranteng P, Karim S, Junaid K, McGuinness B, Passmore AP, Nilforooshan R, Macharouthu A, Donaldson A, Thacker S, Russell G, Malik N, Mate V, Knight L, Kshemendran S, Tan T, Holscher C, Harrison J, Brooks DJ, Ballard C, Edison P. Does insulin resistance influence neurodegeneration in non-diabetic Alzheimer's subjects? ALZHEIMERS RESEARCH & THERAPY 2021; 13:47. [PMID: 33597002 PMCID: PMC7890851 DOI: 10.1186/s13195-021-00784-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 02/03/2021] [Indexed: 12/12/2022]
Abstract
Background Type 2 diabetes is a risk factor for Alzheimer’s disease (AD), and AD brain shows impaired insulin signalling. The role of peripheral insulin resistance on AD aetiopathogenesis in non-diabetic patients is still debated. Here we evaluated the influence of insulin resistance on brain glucose metabolism, grey matter volume and white matter lesions (WMLs) in non-diabetic AD subjects. Methods In total, 130 non-diabetic AD subjects underwent MRI and [18F]FDG PET scans with arterial cannula insertion for radioactivity measurement. T1 Volumetric and FLAIR sequences were acquired on a 3-T MRI scanner. These subjects also had measurement of glucose and insulin levels after a 4-h fast on the same day of the scan. Insulin resistance was calculated by the updated homeostatic model assessment (HOMA2). For [18F]FDG analysis, cerebral glucose metabolic rate (rCMRGlc) parametric images were generated using spectral analysis with arterial plasma input function. Results In this non-diabetic AD population, HOMA2 was negatively associated with hippocampal rCMRGlc, along with total grey matter volumes. No significant correlation was observed between HOMA2, hippocampal volume and WMLs. Conclusions In non-diabetic AD, peripheral insulin resistance is independently associated with reduced hippocampal glucose metabolism and with lower grey matter volume, suggesting that peripheral insulin resistance might influence AD pathology by its action on cerebral glucose metabolism and on neurodegeneration. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00784-w.
Collapse
Affiliation(s)
- Grazia Daniela Femminella
- Division of Neurology, Neurology Imaging Unit, Department of Brain Sciences, Imperial College London, 1st Floor B Block, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Nicholas R Livingston
- Division of Neurology, Neurology Imaging Unit, Department of Brain Sciences, Imperial College London, 1st Floor B Block, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Sanara Raza
- Division of Neurology, Neurology Imaging Unit, Department of Brain Sciences, Imperial College London, 1st Floor B Block, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Thalia van der Doef
- Division of Neurology, Neurology Imaging Unit, Department of Brain Sciences, Imperial College London, 1st Floor B Block, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | | | | | - Gail Busza
- Division of Neurology, Neurology Imaging Unit, Department of Brain Sciences, Imperial College London, 1st Floor B Block, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Valeria Calsolaro
- Division of Neurology, Neurology Imaging Unit, Department of Brain Sciences, Imperial College London, 1st Floor B Block, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Stefan Carver
- Division of Neurology, Neurology Imaging Unit, Department of Brain Sciences, Imperial College London, 1st Floor B Block, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | | | | | - Robert M Lawrence
- South West London and St George's Mental Health NHS Trust, London, UK
| | | | - George Tadros
- Heart of England NHS Foundation Trust, Birmingham, UK
| | - Basil H Ridha
- Brighton and Sussex University Hospital Trust, Brighton, UK
| | | | - Zuzana Walker
- Mental Health Unit, St. Margaret's Hospital, Epping, Essex, UK
| | | | | | - Ben Underwood
- Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
| | - Aparna Prasanna
- Black Country Partnership NHS Foundation Trust, Wolverhampton, UK
| | - Paul Koranteng
- Northamptonshire Healthcare NHS Foundation Trust, Northampton, UK
| | - Salman Karim
- Lancashire Care NHS Foundation Trust, Preston, UK
| | - Kehinde Junaid
- Nottinghamshire Healthcare NHS Foundation Trust, Nottingham, UK
| | | | | | | | | | | | - Simon Thacker
- Derbyshire Healthcare NHS Foundation Trust, Derby, UK
| | - Gregor Russell
- Bradford District Care NHS Foundation Trust, Bradford, UK
| | - Naghma Malik
- North West Boroughs Partnership NHS Foundation Trust, Warrington, UK
| | - Vandana Mate
- Cornwall Partnership NHS Foundation Trust, Redruth, UK
| | - Lucy Knight
- Somerset Partnership NHS Foundation Trust, South Petherton, UK
| | - Sajeev Kshemendran
- South Staffordshire and Shropshire Healthcare NHS Foundation Trust, Shrewsbury, UK
| | - Tricia Tan
- Division of Neurology, Neurology Imaging Unit, Department of Brain Sciences, Imperial College London, 1st Floor B Block, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Christian Holscher
- Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, China
| | - John Harrison
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | | | | - Paul Edison
- Division of Neurology, Neurology Imaging Unit, Department of Brain Sciences, Imperial College London, 1st Floor B Block, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
47
|
Chung SJ, Lee S, Yoo HS, Baik K, Lee HS, Jung JH, Choi Y, Hong JM, Kim YJ, Ye BS, Sohn YH, Yun M, Lee PH. Different patterns of β-amyloid deposition in patients with Alzheimer's disease according to the presence of mild parkinsonism. Neurobiol Aging 2021; 101:199-206. [PMID: 33631471 DOI: 10.1016/j.neurobiolaging.2021.01.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 01/20/2021] [Accepted: 01/26/2021] [Indexed: 10/22/2022]
Abstract
This study aimed to compare the patterns of β-amyloid deposition between patients with early-stage Alzheimer's disease (AD) with mild parkinsonism and those without parkinsonism. Sixty-one patients with early-stage AD (Clinical Dementia Rating [CDR], 0.5 or 1) who underwent 18F-florbetaben (18F-FBB) PET scans were enrolled. We performed comparative analyses of regional FBB uptake in the frontal, parietal, lateral temporal, medial temporal, occipital, anterior cingulate, and posterior cingulate cortices and in the precuneus, striatum, and thalamus between AD patients with mild parkinsonism (AD-p+; n = 23) and those without parkinsonism (AD-p-; n = 38). There was no significant difference in age, sex, years of education, Mini-Mental State Examination score, and white matter hyperintensity severity between groups. The AD-p+ group had lower composite scores in frontal/executive function domain than the AD-p- group. The AD-p+ group had a higher FBB uptake in the occipital cortex, but not in other cortical regions, than the AD-p- group. Our findings suggest that additional β-amyloid deposition in the occipital region is associated with mild parkinsonism in early-stage AD.
Collapse
Affiliation(s)
- Seok Jong Chung
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea; Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, South Korea
| | - Sangwon Lee
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Han Soo Yoo
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - KyoungWon Baik
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Hye Sun Lee
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, South Korea
| | - Jin Ho Jung
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Yonghoon Choi
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji-Man Hong
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea; Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, South Korea
| | - Yun Joong Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea; Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, South Korea
| | - Byoung Seok Ye
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Young H Sohn
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Mijin Yun
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
48
|
Bao YW, Chau ACM, Chiu PKC, Shea YF, Kwan JSK, Chan FHW, Mak HKF. Heterogeneity of Amyloid Binding in Cognitively Impaired Patients Consecutively Recruited from a Memory Clinic: Evaluating the Utility of Quantitative 18F-Flutemetamol PET-CT in Discrimination of Mild Cognitive Impairment from Alzheimer's Disease and Other Dementias. J Alzheimers Dis 2021; 79:819-832. [PMID: 33361593 PMCID: PMC7902948 DOI: 10.3233/jad-200890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND With the more widespread use of 18F-radioligand-based amyloid-β (Aβ) PET-CT imaging, we evaluated Aβ binding and the utility of neocortical 18F-Flutemetamol standardized uptake value ratio (SUVR) as a biomarker. OBJECTIVE 18F-Flutemetamol SUVR was used to differentiate 1) mild cognitive impairment (MCI) from Alzheimer's disease (AD), and 2) MCI from other non-AD dementias (OD). METHODS 109 patients consecutively recruited from a University memory clinic underwent clinical evaluation, neuropsychological test, MRI and 18F-Flutemetamol PET-CT. The diagnosis was made by consensus of a panel consisting of 1 neuroradiologist and 2 geriatricians. The final cohort included 13 subjective cognitive decline (SCD), 22 AD, 39 MCI, and 35 OD. Quantitative analysis of 16 region-of-interests made by Cortex ID software (GE Healthcare). RESULTS The global mean 18F-Flutemetamol SUVR in SCD, MCI, AD, and OD were 0.50 (SD-0.08), 0.53 (SD-0.16), 0.76 (SD-0.10), and 0.56 (SD-0.16), respectively, with SUVR in SCD and MCI and OD being significantly lower than AD. Aβ binding in SCD, MCI, and OD was heterogeneous, being 23%, 38.5%, and 42.9% respectively, as compared to 100% amyloid positivity in AD. Using global SUVR, ROC analysis showed AUC of 0.868 and 0.588 in differentiating MCI from AD and MCI from OD respectively. CONCLUSION 18F-Flutemetamol SUVR differentiated MCI from AD with high efficacy (high negative predictive value), but much lower efficacy from OD. The major benefit of the test was to differentiate cognitively impaired patients (either SCD, MCI, or OD) without AD-related-amyloid-pathology from AD in the clinical setting, which was under-emphasized in the current guidelines proposed by Amyloid Imaging Task Force.
Collapse
Affiliation(s)
- Yi-Wen Bao
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Anson C M Chau
- Department of Medical Imaging, The University of Hong Kong (Shenzhen) Teaching Hospital , The University of Hong Kong, Hong Kong SAR, China
| | - Patrick Ka-Chun Chiu
- Division of Geriatrics, Department of Medicine, Queen Mary Hospital, Hong Kong SAR, China
| | - Yat Fung Shea
- Division of Geriatrics, Department of Medicine, Queen Mary Hospital, Hong Kong SAR, China
| | - Joseph S K Kwan
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Felix Hon Wai Chan
- Division of Geriatrics, Department of Medicine, Queen Mary Hospital, Hong Kong SAR, China
| | - Henry Ka-Fung Mak
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
49
|
Ayton S, Bush AI. β-amyloid: The known unknowns. Ageing Res Rev 2021; 65:101212. [PMID: 33188924 DOI: 10.1016/j.arr.2020.101212] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) stands out as a major disease without any form of preventative or disease modifying therapy. This is not for lack of trying. 33 phase 3 clinical trials of drugs targeting amyloid beta (Aβ) have failed to slow cognitive decline in AD. The field is at a cross-roads about whether to continue anti-Aβ therapy or more actively pursue alternative targets. With the burden of this disease to patients, families, and healthcare budgets growing yearly, the need for disease modifying AD therapies has become one of the highest priorities in all of medicine. While pathology, genetic and biochemical data offer a popular narrative for the causative role of Aβ, there are alternative explanations, and dissenting findings that, now more than ever, warrant thorough reanalysis. This review questions the major assumptions about Aβ on which therapies for AD were premised, and invites renewed interrogation into AD pathogenesis.
Collapse
Affiliation(s)
- Scott Ayton
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia.
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia.
| |
Collapse
|
50
|
Zetterberg H, Bendlin BB. Biomarkers for Alzheimer's disease-preparing for a new era of disease-modifying therapies. Mol Psychiatry 2021; 26:296-308. [PMID: 32251378 PMCID: PMC8172244 DOI: 10.1038/s41380-020-0721-9] [Citation(s) in RCA: 200] [Impact Index Per Article: 66.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/17/2020] [Accepted: 03/24/2020] [Indexed: 12/25/2022]
Abstract
Clinical trial results presented in 2019 suggest that antibody-based removal of cerebral amyloid β (Aβ) plaques may possibly clear tau tangles and modestly slow cognitive decline in symptomatic Alzheimer's disease (AD). Although regulatory approval of this approach is still pending, preparing the healthcare system for the advent of disease-modifying therapies against AD is imperative. In particular, it will be necessary to identify the most suitable biomarkers to facilitate appropriate treatment of AD. Here, we give an update on recent developments in fluid and imaging biomarkers for AD-related pathologies and discuss potential approaches that could be adopted to screen for and clarify the underlying pathology in people seeking medical advice because of cognitive symptoms. We succinctly review recent data regarding biomarkers for Aβ and tau pathology, neurodegeneration, synaptic dysfunction, and inflammation, highlight the need for further research into common copathologies, and suggest how different biomarkers could be used (most likely in combination) to facilitate the development and clinical implementation of novel drug candidates against AD.
Collapse
Affiliation(s)
- Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK.
- UK Dementia Research Institute at UCL, London, UK.
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|