1
|
Wang M, Zhang H, Liang J, Huang J, Wu T, Chen N. Calcium signaling hypothesis: A non-negligible pathogenesis in Alzheimer's disease. J Adv Res 2025:S2090-1232(25)00026-8. [PMID: 39793962 DOI: 10.1016/j.jare.2025.01.007] [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: 08/25/2024] [Revised: 11/23/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) presents a significant challenge to global healthcare systems, with an exacerbation by an aging population. Although the plethora of hypotheses are proposed to elucidate the underlying mechanisms of AD, from amyloid-beta (Aβ) accumulation and Tau protein aggregation to neuroinflammation, a comprehensive understanding of its pathogenesis remains elusive. Recent research has highlighted the critical role of calcium (Ca2+) signaling pathway in the progression of AD, indicating a complex interplay between Ca2+ dysregulation and various pathological processes. AIM OF REVIEW This review aims to consolidate the current understanding of the role of Ca2+ signaling dysregulation in AD, thus emphasizing its central role amidst various pathological hypotheses. We aim to evaluate the potential of the Ca2+ signaling hypothesis to unify existing theories of AD pathogenesis and explore its implications for developing innovative therapeutic strategies through targeting Ca2+ dysregulation. KEY SCIENTIFIC CONCEPTS OF REVIEW The review focuses on three principal concepts. First, the indispensable role of Ca2+ homeostasis in neuronal function and its disruption in AD. Second, the interaction between Ca2+ signaling dysfunction and established AD hypotheses posited that Ca2+ dysregulation is a unifying pathway. Third, the dual role of Ca2+ in neurodegeneration and neuroprotection, highlighting the nuanced effects of Ca2+ levels on AD pathology.
Collapse
Affiliation(s)
- Minghui Wang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Hu Zhang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Jiling Liang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Jielun Huang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Tong Wu
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China.
| | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China.
| |
Collapse
|
2
|
Singh R, Jain S, Paliwal V, Verma K, Paliwal S, Sharma S. Does Metabolic Manager Show Encouraging Outcomes in Alzheimer's?: Challenges and Opportunity for Protein Tyrosine Phosphatase 1b Inhibitors. Drug Dev Res 2024; 85:e70026. [PMID: 39655712 DOI: 10.1002/ddr.70026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/22/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024]
Abstract
Protein tyrosine phosphatase 1b (PTP1b) is a member of the protein tyrosine phosphatase (PTP) enzyme group and encoded as PTP1N gene. Studies have evidenced an overexpression of the PTP1b enzyme in metabolic syndrome, anxiety, schizophrenia, neurodegeneration, and neuroinflammation. PTP1b inhibitor negatively regulates insulin and leptin pathways and has been explored as an antidiabetic agent in various clinical trials. Notably, the preclinical studies have shown that recuperating metabolic dysfunction and dyshomeostasis can reverse cognition and could be a possible approach to mitigate multifaceted Alzheimer's disease (AD). PTP1b inhibitor thus has attracted attention in neuroscience, though the development is limited to the preclinical stage, and its exploration in large clinical trials is warranted. This review provides an insight on the development of PTP1b inhibitors from different sources in diabesity. The crosstalk between metabolic dysfunction and insulin insensitivity in AD and type-2 diabetes has also been highlighted. Furthermore, this review presents the significance of PTP1b inhibition in AD based on pathophysiological facets, and recent evidences from preclinical and clinical studies.
Collapse
Affiliation(s)
- Ritu Singh
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Smita Jain
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Vartika Paliwal
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Kanika Verma
- Department of Internal Medicine, Division of Cardiology, LSU Health Sciences Center Shreveport, Louisiana, USA
| | - Sarvesh Paliwal
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| |
Collapse
|
3
|
Hsiao CP, Goto T, Von Ah D, Saligan LN. Cancer-Related Cognitive Impairment Associated with APOE rs7412 and BDNF rs6265 in Breast Cancer Survivors. Semin Oncol Nurs 2024; 40:151721. [PMID: 39198096 DOI: 10.1016/j.soncn.2024.151721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 09/01/2024]
Abstract
OBJECTIVES Cancer-related cognitive impairment (CRCI) is a highly prevalent and debilitating symptom reported by breast cancer survivors (BCS). The etiology of CRCI remains unclear, leading to poor symptom management. Building from prior studies, BCS with the C/C genotype of apolipoprotein E (APOE) rs7412 and the T/T genotype of brain-derived neurotrophic factor (BDNF) rs6265 were hypothesized to experience more severe CRCI. Therefore, we investigated the relationships between the severity of CRCI and polymorphisms of APOE and BDNF among BCS. METHODS This was a subanalysis of data from a larger descriptive, correlational, and cross-sectional study. Subjective and objective CRCI were measured using the Patient-Reported Outcomes Measurement Information System and CANTAB Cambridge Cognitive assessment, respectively. Buccal swab samples were collected to evaluate the single nucleotide polymorphisms. Multivariable generalized linear regression models were used to analyze data. RESULTS APOE rs7412 and BDNF rs6265 were significantly associated with lower self-reported cognitive abilities in a total of 353 BCS. Age was positively associated with self-reported cognitive scores, indicating that younger BCS perceived lower cognitive abilities. Individuals carrying genotype of C/T for APOE with the C/C or C/T for BDNF showed positive associations with cognitive abilities. CONCLUSIONS Younger BCS with the C/C genotype for APOE rs7412 and the T/T genotype for BDNF rs6265 may be at risk for CRCI. Knowledge regarding predictive markers for CRCI symptoms is essential for precision symptom management. Further investigation with a longitudinal and translational design is necessary to explore the etiologies for CRCI. IMPLICATIONS FOR NURSING PRACTICE Integrating genetic phenotyping into routine clinical practice will provide nurses with unique opportunities to understand individual susceptibilities, and how symptoms may trigger other symptoms. Further, findings from these innovative investigations will provide symptom interventionists and implementation scientists with critical data to optimize individualized strategies for symptom prevention, detection, and management.
Collapse
Affiliation(s)
- Chao-Pin Hsiao
- Case Western Reserve University School of Nursing, Cleveland, Ohio
| | - Taichi Goto
- Symptoms Biology Unit, Division of Intramural Research, National Institute of Nursing Research, National Institutes of Health, Bethesda, Maryland
| | - Diane Von Ah
- The Ohio State University College of Nursing, Columbus, Ohio
| | - Leorey N Saligan
- Symptoms Biology Unit, Division of Intramural Research, National Institute of Nursing Research, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
4
|
Moawad MHED, Serag I, Alkhawaldeh IM, Abbas A, Sharaf A, Alsalah S, Sadeq MA, Shalaby MMM, Hefnawy MT, Abouzid M, Meshref M. Exploring the Mechanisms and Therapeutic Approaches of Mitochondrial Dysfunction in Alzheimer's Disease: An Educational Literature Review. Mol Neurobiol 2024:10.1007/s12035-024-04468-y. [PMID: 39254911 DOI: 10.1007/s12035-024-04468-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: 01/30/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024]
Abstract
Alzheimer's disease (AD) presents a significant challenge to global health. It is characterized by progressive cognitive deterioration and increased rates of morbidity and mortality among older adults. Among the various pathophysiologies of AD, mitochondrial dysfunction, encompassing conditions such as increased reactive oxygen production, dysregulated calcium homeostasis, and impaired mitochondrial dynamics, plays a pivotal role. This review comprehensively investigates the mechanisms of mitochondrial dysfunction in AD, focusing on aspects such as glucose metabolism impairment, mitochondrial bioenergetics, calcium signaling, protein tau and amyloid-beta-associated synapse dysfunction, mitophagy, aging, inflammation, mitochondrial DNA, mitochondria-localized microRNAs, genetics, hormones, and the electron transport chain and Krebs cycle. While lecanemab is the only FDA-approved medication to treat AD, we explore various therapeutic modalities for mitigating mitochondrial dysfunction in AD, including antioxidant drugs, antidiabetic agents, acetylcholinesterase inhibitors (FDA-approved to manage symptoms), nutritional supplements, natural products, phenylpropanoids, vaccines, exercise, and other potential treatments.
Collapse
Affiliation(s)
- Mostafa Hossam El Din Moawad
- Faculty of Pharmacy, Clinical Department, Alexandria Main University Hospital, Alexandria, Egypt
- Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Ibrahim Serag
- Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | | | - Abdallah Abbas
- Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| | - Abdulrahman Sharaf
- Department of Clinical Pharmacy, Salmaniya Medical Complex, Government Hospital, Manama, Bahrain
| | - Sumaya Alsalah
- Ministry of Health, Primary Care, Governmental Health Centers, Manama, Bahrain
| | | | | | | | - Mohamed Abouzid
- Department of Physical Pharmacy and Pharmacokinetics, Faculty of Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3 St., 60-806, Poznan, Poland.
- Doctoral School, Poznan University of Medical Sciences, 60-812, Poznan, Poland.
| | - Mostafa Meshref
- Department of Neurology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
5
|
Chen F, Zhao J, Meng F, He F, Ni J, Fu Y. The vascular contribution of apolipoprotein E to Alzheimer's disease. Brain 2024; 147:2946-2965. [PMID: 38748848 DOI: 10.1093/brain/awae156] [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: 12/24/2023] [Revised: 03/23/2024] [Accepted: 04/21/2024] [Indexed: 09/04/2024] Open
Abstract
Alzheimer's disease, the most prevalent form of dementia, imposes a substantial societal burden. The persistent inadequacy of disease-modifying drugs targeting amyloid plaques and neurofibrillary tangles suggests the contribution of alternative pathogenic mechanisms. A frequently overlooked aspect is cerebrovascular dysfunction, which may manifest early in the progression of Alzheimer's disease pathology. Mounting evidence underscores the pivotal role of the apolipoprotein E gene, particularly the apolipoprotein ε4 allele as the strongest genetic risk factor for late-onset Alzheimer's disease, in the cerebrovascular pathology associated with Alzheimer's disease. In this review, we examine the evidence elucidating the cerebrovascular impact of both central and peripheral apolipoprotein E on the pathogenesis of Alzheimer's disease. We present a novel three-hit hypothesis, outlining potential mechanisms that shed light on the intricate relationship among different pathogenic events. Finally, we discuss prospective therapeutics targeting the cerebrovascular pathology associated with apolipoprotein E and explore their implications for future research endeavours.
Collapse
Affiliation(s)
- Feng Chen
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jing Zhao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Fanxia Meng
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Fangping He
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jie Ni
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuan Fu
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
6
|
Leclerc M, Tremblay C, Bourassa P, Schneider JA, Bennett DA, Calon F. Lower GLUT1 and unchanged MCT1 in Alzheimer's disease cerebrovasculature. J Cereb Blood Flow Metab 2024; 44:1417-1432. [PMID: 38441044 PMCID: PMC11342728 DOI: 10.1177/0271678x241237484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 12/21/2023] [Accepted: 01/16/2024] [Indexed: 03/06/2024]
Abstract
The brain is a highly demanding organ, utilizing mainly glucose but also ketone bodies as sources of energy. Glucose transporter-1 (GLUT1) and monocarboxylates transporter-1 (MCT1) respectively transport glucose and ketone bodies across the blood-brain barrier. While reduced glucose uptake by the brain is one of the earliest signs of Alzheimer's disease (AD), no change in the uptake of ketone bodies has been evidenced yet. To probe for changes in GLUT1 and MCT1, we performed Western immunoblotting in microvessel extracts from the parietal cortex of 60 participants of the Religious Orders Study. Participants clinically diagnosed with AD had lower cerebrovascular levels of GLUT1, whereas MCT1 remained unchanged. GLUT1 reduction was associated with lower cognitive scores. No such association was found for MCT1. GLUT1 was inversely correlated with neuritic plaques and cerebrovascular β-secretase-derived fragment levels. No other significant associations were found between both transporters, markers of Aβ and tau pathologies, sex, age at death or apolipoprotein-ε4 genotype. These results suggest that, while a deficit of GLUT1 may underlie the reduced transport of glucose to the brain in AD, no such impairment occurs for MCT1. This study thus supports the exploration of ketone bodies as an alternative energy source for the aging brain.
Collapse
Affiliation(s)
- Manon Leclerc
- Faculté de pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec – Université Laval, Québec, Canada
| | - Cyntia Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec – Université Laval, Québec, Canada
| | - Philippe Bourassa
- Faculté de pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec – Université Laval, Québec, Canada
| | - Julie A Schneider
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Frédéric Calon
- Faculté de pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec – Université Laval, Québec, Canada
| |
Collapse
|
7
|
Shah SN, Dounavi ME, Malhotra PA, Lawlor B, Naci L, Koychev I, Ritchie CW, Ritchie K, O’Brien JT. Dementia risk and thalamic nuclei volumetry in healthy midlife adults: the PREVENT Dementia study. Brain Commun 2024; 6:fcae046. [PMID: 38444908 PMCID: PMC10914447 DOI: 10.1093/braincomms/fcae046] [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: 07/26/2023] [Revised: 12/31/2023] [Accepted: 02/13/2024] [Indexed: 03/07/2024] Open
Abstract
A reduction in the volume of the thalamus and its nuclei has been reported in Alzheimer's disease, mild cognitive impairment and asymptomatic individuals with risk factors for early-onset Alzheimer's disease. Some studies have reported thalamic atrophy to occur prior to hippocampal atrophy, suggesting thalamic pathology may be an early sign of cognitive decline. We aimed to investigate volumetric differences in thalamic nuclei in middle-aged, cognitively unimpaired people with respect to dementia family history and apolipoprotein ε4 allele carriership and the relationship with cognition. Seven hundred participants aged 40-59 years were recruited into the PREVENT Dementia study. Individuals were stratified according to dementia risk (approximately half with and without parental dementia history). The subnuclei of the thalamus of 645 participants were segmented on T1-weighted 3 T MRI scans using FreeSurfer 7.1.0. Thalamic nuclei were grouped into six regions: (i) anterior, (ii) lateral, (iii) ventral, (iv) intralaminar, (v) medial and (vi) posterior. Cognitive performance was evaluated using the computerized assessment of the information-processing battery. Robust linear regression was used to analyse differences in thalamic nuclei volumes and their association with cognitive performance, with age, sex, total intracranial volume and years of education as covariates and false discovery rate correction for multiple comparisons. We did not find significant volumetric differences in the thalamus or its subregions, which survived false discovery rate correction, with respect to first-degree family history of dementia or apolipoprotein ε4 allele status. Greater age was associated with smaller volumes of thalamic subregions, except for the medial thalamus, but only in those without a dementia family history. A larger volume of the mediodorsal medial nucleus (Pfalse discovery rate = 0.019) was associated with a faster processing speed in those without a dementia family history. Larger volumes of the thalamus (P = 0.016) and posterior thalamus (Pfalse discovery rate = 0.022) were associated with significantly worse performance in the immediate recall test in apolipoprotein ε4 allele carriers. We did not find significant volumetric differences in thalamic subregions in relation to dementia risk but did identify an interaction between dementia family history and age. Larger medial thalamic nuclei may exert a protective effect on cognitive performance in individuals without a dementia family history but have little effect on those with a dementia family history. Larger volumes of posterior thalamic nuclei were associated with worse recall in apolipoprotein ε4 carriers. Our results could represent initial dysregulation in the disease process; further study is needed with functional imaging and longitudinal analysis.
Collapse
Affiliation(s)
- Sita N Shah
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Maria-Eleni Dounavi
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Paresh A Malhotra
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK
- UK Dementia Research Institute Care Research and Technology Centre, Imperial College London and the University of Surrey, London SW7 2AZ, UK
| | - Brian Lawlor
- Trinity College Institute of Neuroscience, School of Psychology, Trinity College Dublin, Dublin D02 PX31, Ireland
- Global Brain Health Institute, Trinity College Dublin, Dublin D02 X9W9, Ireland
| | - Lorina Naci
- Trinity College Institute of Neuroscience, School of Psychology, Trinity College Dublin, Dublin D02 PX31, Ireland
- Global Brain Health Institute, Trinity College Dublin, Dublin D02 X9W9, Ireland
| | - Ivan Koychev
- Department of Psychiatry, University of Oxford, Oxford OX3 7JX, UK
| | - Craig W Ritchie
- Centre for Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Karen Ritchie
- Institute de Neurosciences de Montpellier, INSERM, Montpellier 34093, France
| | - John T O’Brien
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| |
Collapse
|
8
|
Chen G, McKay NS, Gordon BA, Liu J, Joseph-Mathurin N, Schindler SE, Hassenstab J, Aschenbrenner AJ, Wang Q, Schultz SA, Su Y, LaMontagne PJ, Keefe SJ, Massoumzadeh P, Cruchaga C, Xiong C, Morris JC, Benzinger TLS. Predicting cognitive decline: Which is more useful, baseline amyloid levels or longitudinal change? Neuroimage Clin 2023; 41:103551. [PMID: 38150745 PMCID: PMC10788301 DOI: 10.1016/j.nicl.2023.103551] [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: 12/08/2023] [Accepted: 12/10/2023] [Indexed: 12/29/2023]
Abstract
The use of biomarkers for the early detection of Alzheimer's disease (AD) is crucial for developing potential therapeutic treatments. Positron Emission Tomography (PET) is a well-established tool used to detect β-amyloid (Aβ) plaques in the brain. Previous studies have shown that cross-sectional biomarkers can predict cognitive decline (Schindler et al.,2021). However, it is still unclear whether longitudinal Aβ-PET may have additional value for predicting time to cognitive impairment in AD. The current study aims to evaluate the ability of baseline- versus longitudinal rate of change in-11C-Pittsburgh compound B (PiB) Aβ-PET to predict cognitive decline. A cohort of 153 participants who previously underwent PiB-PET scans and comprehensive clinical assessments were used in this study. Our analyses revealed that baseline Aβ is significantly associated with the rate of change in cognitive composite scores, with cognition declining more rapidly when baseline PiB Aβ levels were higher. In contrast, no signification association was identified between the rate of change in PiB-PET Aβ and cognitive decline. Additionally, the ability of the rate of change in the PiB-PET measures to predict cognitive decline was significantly influenced by APOE ε4 carrier status. These results suggest that a single PiB-PET scan is sufficient to predict cognitive decline and that longitudinal measures of Aβ accumulation do not improve the prediction of cognitive decline once someone is amyloid positive.
Collapse
Affiliation(s)
- Gengsheng Chen
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Nicole S McKay
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Jingxia Liu
- Department of Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Nelly Joseph-Mathurin
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Andrew J Aschenbrenner
- Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Qing Wang
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Stephanie A Schultz
- Department of Neurology Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yi Su
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Pamela J LaMontagne
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Sarah J Keefe
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Parinaz Massoumzadeh
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Chengjie Xiong
- Divison of Biostatistics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
9
|
Perovnik M, Tang CC, Namías M, Eidelberg D. Longitudinal changes in metabolic network activity in early Alzheimer's disease. Alzheimers Dement 2023; 19:4061-4072. [PMID: 37204815 DOI: 10.1002/alz.13137] [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: 02/23/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/20/2023]
Abstract
INTRODUCTION The progression of Alzheimer's disease (AD) has been linked to two metabolic networks, the AD-related pattern (ADRP) and the default mode network (DMN). METHODS Converting and clinically stable cognitively normal subjects (n = 47) and individuals with mild cognitive impairment (n = 96) underwent 2-[18 F]fluoro-2-deoxy-D-glucose (FDG) positron emission tomography (PET) three or more times over 6 years (nscans = 705). Expression levels for ADRP and DMN were measured in each subject and time point, and the resulting changes were correlated with cognitive performance. The role of network expression in predicting conversion to dementia was also evaluated. RESULTS Longitudinal increases in ADRP expression were observed in converters, while age-related DMN loss was seen in converters and nonconverters. Cognitive decline correlated with increases in ADRP and declines in DMN, but conversion to dementia was predicted only by baseline ADRP levels. DISCUSSION The results point to the potential utility of ADRP as an imaging biomarker of AD progression.
Collapse
Affiliation(s)
- Matej Perovnik
- Department of Neurology, University Medical Center Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Chris C Tang
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Mauro Namías
- Fundación Centro Diagnóstico Nuclear, Buenos Aires, Argentina
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Molecular Medicine and Neurology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| |
Collapse
|
10
|
Kapadia A, Billimoria K, Desai P, Grist JT, Heyn C, Maralani P, Symons S, Zaccagna F. Hypoperfusion Precedes Tau Deposition in the Entorhinal Cortex: A Retrospective Evaluation of ADNI-2 Data. J Clin Neurol 2023; 19:131-137. [PMID: 36647226 PMCID: PMC9982189 DOI: 10.3988/jcn.2022.0088] [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: 02/24/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND AND PURPOSE Tau deposition in the entorhinal cortex is the earliest pathological feature of Alzheimer's disease (AD). However, this feature has also been observed in cognitively normal (CN) individuals and those with mild cognitive impairment (MCI). The precise pathophysiology for the development of tau deposition remains unclear. We hypothesized that reduced cerebral perfusion is associated with the development of tau deposition. METHODS A subset of the Alzheimer's Disease Neuroimaging Initiative data set was utilized. Included patients had undergone arterial spin labeling perfusion MRI along with [18F]flortaucipir tau PET at baseline, within 1 year of the MRI, and a follow-up at 6 years. The association between baseline cerebral blood flow (CBF) and the baseline and 6-year tau PET was assessed. Univariate and multivariate linear modeling was performed, with p<0.05 indicating significance. RESULTS Significant differences were found in the CBF between patients with AD and MCI, and CN individuals in the left entorhinal cortex (p=0.013), but not in the right entorhinal cortex (p=0.076). The difference in maximum standardized uptake value ratio between 6 years and baseline was significantly and inversely associated with the baseline mean CBF (p=0.042, R²=0.54) in the left entorhinal cortex but not the right entorhinal cortex. Linear modeling demonstrated that CBF predicted 6-year tau deposition (p=0.015, R²=0.11). CONCLUSIONS The results of this study suggest that a reduction in CBF at the entorhinal cortex precedes tau deposition. Further work is needed to understand the mechanism underlying tau deposition in aging and disease.
Collapse
Affiliation(s)
- Anish Kapadia
- Division of Neuroradiology, Department of Medical Imaging, University of Toronto, Toronto, ON, Canada.,Division of Neuroradiology, Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.
| | - Krish Billimoria
- MD Program, Temetry Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Prarthna Desai
- Department of Medicine, Maharaja Sayajirao University of Baroda, Vadodara, India
| | - James T. Grist
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, UK.,Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, UK.,Department of Radiology, Oxford University Hospitals Trust, Oxford, UK.,Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Chris Heyn
- Division of Neuroradiology, Department of Medical Imaging, University of Toronto, Toronto, ON, Canada.,Division of Neuroradiology, Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Pejman Maralani
- Division of Neuroradiology, Department of Medical Imaging, University of Toronto, Toronto, ON, Canada.,Division of Neuroradiology, Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Sean Symons
- Division of Neuroradiology, Department of Medical Imaging, University of Toronto, Toronto, ON, Canada.,Division of Neuroradiology, Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Fulvio Zaccagna
- Division of Neuroradiology, Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
11
|
Li W, Li R, Yan S, Zhao Z, Shan Y, Qi Z, Lu J. Effect of APOE ε4 genotype on amyloid-β, glucose metabolism, and gray matter volume in cognitively normal individuals and amnestic mild cognitive impairment. Eur J Neurol 2023; 30:587-596. [PMID: 36448771 PMCID: PMC10107141 DOI: 10.1111/ene.15656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND AND PURPOSE The presence of apolipoprotein E ε4 (APOE ε4) is associated with an increased risk of developing Alzheimer disease (AD). The aim of this study was to assess the effects of APOE ε4 on amyloid-β (Aβ) pathology, glucose metabolism, and gray matter (GM) volume and their longitudinal changes in healthy control (HC) and amnestic mild cognitive impairment (aMCI). METHODS We included 50 HCs and 109 aMCI patients from the Alzheimer's Disease Neuroimaging Initiative phase 2/GO based on availability of baseline T1-weighted magnetic resonance imaging, 18 F-florbetapir positron emission tomography (PET), and 18 F-fluorodeoxyglucose (FDG) PET. Of these, 35 HCs and 67 aMCI patients who underwent 24-month scans were included for follow-up study. RESULTS Voxelwise analysis revealed that APOE ε4 carriers exhibited greater baseline Aβ deposition than APOE ε4 noncarriers in both diagnostic groups. However, there was no significant difference between APOE ε4 noncarriers and APOE ε4 carriers in terms of 18 F-FDG PET standardized uptake value ratio and GM volume. Region of interest-based analysis showed statistically significant greater Aβ deposition in APOE ε4 carriers than APOE ε4 noncarriers only in aMCI patients. Furthermore, APOE ε4 carriers generally exhibited a greater magnitude and spatial extent of longitudinal changes in Aβ deposition than APOE ε4 noncarriers in both diagnostic groups. CONCLUSIONS Our findings suggest a differential effect of APOE ε4 on Aβ pathology, glucose metabolism, and GM volume. Studying APOE ε4-related brain changes with neuroimaging biomarkers in preclinical AD offers an opportunity to further our understanding of the pathophysiology of AD at an early stage.
Collapse
Affiliation(s)
- Weihua Li
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | | | - Shaozhen Yan
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Zhilian Zhao
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Yi Shan
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Zhigang Qi
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | | |
Collapse
|
12
|
de Frutos Lucas J, Sewell KR, García-Colomo A, Markovic S, Erickson KI, Brown BM. How does apolipoprotein E genotype influence the relationship between physical activity and Alzheimer's disease risk? A novel integrative model. Alzheimers Res Ther 2023; 15:22. [PMID: 36707869 PMCID: PMC9881295 DOI: 10.1186/s13195-023-01170-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 01/15/2023] [Indexed: 01/29/2023]
Abstract
BACKGROUND Wide evidence suggests that physical activity (PA) confers protection against Alzheimer's disease (AD). On the other hand, the apolipoprotein E gene (APOE) ε4 allele represents the greatest genetic risk factor for developing AD. Extensive research has been conducted to determine whether frequent PA can mitigate the increased AD risk associated with APOE ε4. However, thus far, these attempts have produced inconclusive results. In this context, one possible explanation could be that the influence of the combined effect of PA and APOE ε4 carriage might be dependent on the specific outcome measure utilised. MAIN BODY In order to bridge these discrepancies, the aim of this theoretical article is to propose a novel model on the interactive effects of PA and APOE ε4 carriage on well-established mechanisms underlying AD. Available literature was searched to investigate how PA and APOE ε4 carriage, independently and in combination, may alter several molecular pathways involved in AD pathogenesis. The reviewed mechanisms include amyloid beta (Aβ) and tau deposition and clearance, neuronal resilience and neurogenesis, lipid function and cerebrovascular alterations, brain immune response and glucose metabolism. Finally, combining all this information, we have built an integrative model, which includes evidence-based and theoretical synergistic interactions across mechanisms. Moreover, we have identified key knowledge gaps in the literature, providing a list of testable hypotheses that future studies need to address. CONCLUSIONS We conclude that PA influences a wide array of molecular targets involved in AD neuropathology. A deeper understanding of where, when and, most importantly, how PA decreases AD risk even in the presence of the APOE ε4 allele will enable the creation of new protocols using exercise along pharmaceuticals in combined therapeutic approaches.
Collapse
Affiliation(s)
- Jaisalmer de Frutos Lucas
- Experimental Psychology, Cognitive Processes and Logopedia Department, School of Psychology, Universidad Complutense de Madrid, 28223, Pozuelo de Alarcón, Spain.
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, 6027, Australia.
- Departamento de PsicologíaFacultad de Ciencias de la Vida y de la Naturaleza, Universidad Antonio de Nebrija, 28015, Madrid, Spain.
| | - Kelsey R Sewell
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, 6150, Australia
| | - Alejandra García-Colomo
- Experimental Psychology, Cognitive Processes and Logopedia Department, School of Psychology, Universidad Complutense de Madrid, 28223, Pozuelo de Alarcón, Spain
| | - Shaun Markovic
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, 6150, Australia
- Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, 6009, Australia
| | - Kirk I Erickson
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, 15260, USA
- PROFITH "PROmoting FITness and Health Through Physical Activity" Research Group, Sport and Health University Research Institute (iMUDS), Department of Physical and Sports Education, Faculty of Sport Sciences, University of Granada, 18071, Granada, Spain
- AdventHealth Research Institute, Orlando, FL, 32804, USA
| | - Belinda M Brown
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, 6027, Australia
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, 6150, Australia
- Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, 6009, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia
| |
Collapse
|
13
|
Linear Mixed Model Analysis of Polygenic Hazard Score on Verbal Memory Decline in Alzheimer's Disease. Nurs Res 2023; 72:66-73. [PMID: 36097266 DOI: 10.1097/nnr.0000000000000623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a chronic, progressive, degenerative disease characterized by cognitive dysfunction, including verbal memory loss. Studies were lacking in examining the longitudinal effect of polygenic hazard score on the Rey Auditory Verbal Learning Test-Delayed Total (AVDELTOT) score (a common measure of verbal memory). A key step in analyzing longitudinal changes in cognitive measures using a linear mixed model (LMM) is choosing a suitable covariance structure. OBJECTIVES The study aims to determine the association between the polygenic hazard score and the AVDELTOT score accounting for repeated measures (the covariance structure). METHODS The AVDELTOT scores were collected at baseline, 12 months, 24 months, 36 months, and 48 months from 283 participants with AD, 347 with cognitive normal, and 846 with mild cognitive impairment in the Alzheimer's Disease Neuroimaging Initiative. The Bayesian information criterion statistic was used to select the best covariance structure from 10 covariance structures in longitudinal analysis of AVDELTOT scores. The multivariable LMM was used to investigate the effect of polygenic hazard score status (low vs. medium vs. high) on changes in AVDELTOT scores while adjusted for age, gender, education, APOE-ε4 genotype, and baseline Mini-Mental State Examination score. RESULTS One-way analysis of variance revealed significant differences in AVDELTOT scores, Mini-Mental State Examination scores, and polygenic hazard scores among AD diagnoses at baseline. Bayesian information criterion favored the compound symmetry covariance structure in the LMM analysis. Using the multivariate LMM, the APOE-ε4 allele and high polygenic hazard score value was significantly associated with AVDELTOT declines. Significant polygenic hazard score status by follow-up visit interactions was discovered. CONCLUSION Our findings provide the first evidence of the effect of polygenic hazard score status and APOE-ε4 allele on declines in verbal memory in people with AD.
Collapse
|
14
|
Myette-Côté É, Soto-Mota A, Cunnane SC. Ketones: potential to achieve brain energy rescue and sustain cognitive health during ageing. Br J Nutr 2022; 128:407-423. [PMID: 34581265 DOI: 10.1017/s0007114521003883] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
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
Affiliation(s)
- Étienne Myette-Côté
- Montreal Clinical Research Institute, Montreal, QC, Canada
- Department of Medicine, McGill University, Montreal, QC, Canada
| | - Adrian Soto-Mota
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Stephen C Cunnane
- Research Center on Aging, CIUSSS de l'Estrie - CHUS, Sherbrooke, QC, Canada
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
- Department of Pharmacology & Physiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
15
|
Li Y, Ng YL, Paranjpe MD, Ge Q, Gu F, Li P, Yan S, Lu J, Wang X, Zhou Y. Tracer-specific reference tissues selection improves detection of 18 F-FDG, 18 F-florbetapir, and 18 F-flortaucipir PET SUVR changes in Alzheimer's disease. Hum Brain Mapp 2022; 43:2121-2133. [PMID: 35165964 PMCID: PMC8996354 DOI: 10.1002/hbm.25774] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/17/2021] [Accepted: 12/30/2021] [Indexed: 01/05/2023] Open
Abstract
This study sought to identify a reference tissue‐based quantification approach for improving the statistical power in detecting changes in brain glucose metabolism, amyloid, and tau deposition in Alzheimer's disease studies. A total of 794, 906, and 903 scans were included for 18F‐FDG, 18F‐florbetapir, and 18F‐flortaucipir, respectively. Positron emission tomography (PET) and T1‐weighted images of participants were collected from the Alzheimer's disease Neuroimaging Initiative database, followed by partial volume correction. The standardized uptake value ratios (SUVRs) calculated from the cerebellum gray matter, centrum semiovale, and pons were evaluated at both region of interest (ROI) and voxelwise levels. The statistical power of reference tissues in detecting longitudinal SUVR changes was assessed via paired t‐test. In cross‐sectional analysis, the impact of reference tissue‐based SUVR differences between cognitively normal and cognitively impaired groups was evaluated by effect sizes Cohen's d and two sample t‐test adjusted by age, sex, and education levels. The average ROI t values of pons were 86.62 and 38.40% higher than that of centrum semiovale and cerebellum gray matter in detecting glucose metabolism decreases, while the centrum semiovale reference tissue‐based SUVR provided higher t values for the detection of amyloid and tau deposition increases. The three reference tissues generated comparable d images for 18F‐FDG, 18F‐florbetapir, and 18F‐flortaucipir and comparable t maps for 18F‐florbetapir and 18F‐flortaucipir, but pons‐based t map showed superior performance in 18F‐FDG. In conclusion, the tracer‐specific reference tissue improved the detection of 18F‐FDG, 18F‐florbetapir, and 18F‐flortaucipir PET SUVR changes, which helps the early diagnosis, monitoring of disease progression, and therapeutic response in Alzheimer's disease.
Collapse
Affiliation(s)
- Yanxiao Li
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China.,School of Computer Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Yee Ling Ng
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China
| | - Manish D Paranjpe
- Harvard-MIT Health Sciences and Technology Program, Harvard Medical School, Boston, Massachusetts, USA
| | - Qi Ge
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China
| | - Fengyun Gu
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China.,Department of Statistics, University College Cork, Cork, Ireland
| | - Panlong Li
- School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, Henan, China
| | - Shaozhen Yan
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiuying Wang
- School of Computer Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Yun Zhou
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China
| | | |
Collapse
|
16
|
Clarke H, Messaritaki E, Dimitriadis SI, Metzler-Baddeley C. Dementia Risk Factors Modify Hubs but Leave Other Connectivity Measures Unchanged in Asymptomatic Individuals: A Graph Theoretical Analysis. Brain Connect 2022; 12:26-40. [PMID: 34030485 PMCID: PMC8867081 DOI: 10.1089/brain.2020.0935] [Citation(s) in RCA: 6] [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] [Indexed: 01/12/2023] Open
Abstract
Background: Alzheimer's disease (AD) is the most common form of dementia with genetic and environmental risk contributing to its development. Graph theoretical analyses of brain networks constructed from structural and functional magnetic resonance imaging (MRI) measurements have identified connectivity changes in AD and individuals with mild cognitive impairment. However, brain connectivity in asymptomatic individuals at risk of AD remains poorly understood. Methods: We analyzed diffusion-weighted MRI data from 161 asymptomatic individuals (38-71 years) from the Cardiff Ageing and Risk of Dementia Study (CARDS). We calculated white matter tracts and constructed whole-brain, default mode network (DMN) and visual structural brain networks that incorporate multiple structural metrics as edge weights. We then calculated the relationship of three AD risk factors, namely Apolipoprotein-E ɛ4 (APOE4) genotype, family history of dementia (FH), and central obesity (Waist-Hip-Ratio [WHR]), on graph theoretical measures and hubs. Results: We observed no risk-related differences in clustering coefficients, characteristic path lengths, eccentricity, diameter, and radius across the whole-brain, DMN or visual system. However, a hub in the right paracentral lobule was present in all the high-risk groups (FH, APOE4, obese), but absent in low-risk groups (no FH, APOE4-ve, healthy WHR). Discussion: We identified no risk-related effects on graph theoretical metrics in the structural brain networks of cognitively healthy individuals. However, high risk was associated with a hub in the right paracentral lobule, a medial fronto-parietal cortical area with motor and sensory functions. This finding is consistent with accumulating evidence for right parietal cortex contributions in AD. If this phenotype is shown to predict symptom development in longitudinal studies, it could be used as an early biomarker of AD. Impact statement Alzheimer's disease (AD) is a common form of dementia that to date has no cure. Identifying early biomarkers will aid the discovery and development of treatments that may slow AD progression in the future. In this article, we report that asymptomatic individuals at heightened risk of dementia due to their family history, Apolipoprotein-E ɛ4 genotype, and central adiposity have a hub in the right paracentral lobule, which is absent in low-risk groups. If this phenotype were to predict the development of symptoms in a longitudinal study of the same cohort, it could provide an early biomarker of disease progression.
Collapse
Affiliation(s)
- Hannah Clarke
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, United Kingdom
- School of Medicine, UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Eirini Messaritaki
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, United Kingdom
- BRAIN Biomedical Research Unit, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Stavros I. Dimitriadis
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, United Kingdom
- Neuroinformatics Group, Cardiff University Brain Research Imaging Centre, School of Psychology, Cardiff University, Cardiff, United Kingdom
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
- School of Psychology, Cardiff University, Cardiff, United Kingdom
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff, United Kingdom
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Claudia Metzler-Baddeley
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, United Kingdom
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
17
|
18F-FDG-PET correlates of aging and disease course in ALS as revealed by distinct PVC approaches. Eur J Radiol Open 2022; 9:100394. [PMID: 35059473 PMCID: PMC8760536 DOI: 10.1016/j.ejro.2022.100394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/23/2021] [Accepted: 01/06/2022] [Indexed: 11/23/2022] Open
|
18
|
Age, sex and APOE-ε4 modify the balance between soluble and fibrillar β-amyloid in non-demented individuals: topographical patterns across two independent cohorts. Mol Psychiatry 2022; 27:2010-2018. [PMID: 35236958 PMCID: PMC9126807 DOI: 10.1038/s41380-022-01436-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 12/20/2021] [Accepted: 01/04/2022] [Indexed: 12/16/2022]
Abstract
Amyloid (Aβ) pathology is the earliest detectable pathophysiological event along the Alzheimer's continuum, which can be measured both in the cerebrospinal fluid (CSF) and by Positron Emission Tomography (PET). Yet, these biomarkers identify two distinct Aβ pools, reflecting the clearance of soluble Aβ as opposed to the presence of Aβ fibrils in the brain. An open question is whether risk factors known to increase Alzheimer's' disease (AD) prevalence may promote an imbalance between soluble and deposited Aβ. Unveiling such interactions shall aid our understanding of the biological pathways underlying Aβ deposition and foster the design of effective prevention strategies. We assessed the impact of three major AD risk factors, such as age, APOE-ε4 and female sex, on the association between CSF and PET Aβ, in two independent samples of non-demented individuals (ALFA: n = 320, ADNI: n = 682). We tested our hypotheses both in candidate regions of interest and in the whole brain using voxel-wise non-parametric permutations. All of the assessed risk factors induced a higher Aβ deposition for any given level of CSF Aβ42/40, although in distinct cerebral topologies. While age and sex mapped onto neocortical areas, the effect of APOE-ε4 was prominent in the medial temporal lobe, which represents a target of early tau deposition. Further, we found that the effects of age and APOE-ε4 was stronger in women than in men. Our data indicate that specific AD risk factors affect the spatial patterns of cerebral Aβ aggregation, with APOE-ε4 possibly facilitating a co-localization between Aβ and tau along the disease continuum.
Collapse
|
19
|
Li J, Zheng C, Ge Q, Yan S, Paranjpe MD, Hu S, Zhou Y. Association between long-term donepezil treatment and brain regional amyloid and tau burden among individuals with mild cognitive impairment assessed using 18 F-AV-45 and 18 F-AV-1451 PET. J Neurosci Res 2021; 100:670-680. [PMID: 34882830 DOI: 10.1002/jnr.24995] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 10/16/2021] [Accepted: 11/06/2021] [Indexed: 11/06/2022]
Abstract
This study aims to investigate the association between long-term donepezil treatment and brain neuropathological burden and cognitive function in mild cognitive impairment (MCI) patients. Preprocessed 18 F-AV-45 amyloid and 18 F-AV-1451 tau positron emission tomography (PET) images, magnetic resonance imaging images (MRIs), demographic information, and donepezil use status were downloaded from 255 MCI participants enrolled in the Alzheimer's Disease Neuroimaging Initiative database. Partial volume correction was applied to all PET images. Structural MRIs were used for PET spatial normalization. Regions of interest (ROIs) were defined in standard space, and standardized uptake value ratio (SUVR) images relative to the cerebellum were computed. Multiple linear regression with the least absolute shrinkage selector operator was performed to analyze the effect of long-term donepezil treatment on (a) the SUVR of each 18 F-AV-45 or 18 F-AV-1451 brain PET ROI after adjusting for age, sex, education, ApoE ε4 status, and AD-associated disease risk factors; and (b) cognitive performance after adjusting for age, sex education, ApoE ε4 status, AD-associated disease risk factors, and regional amyloid or tau burden. In adjusted models, long-term donepezil treatment was associated with greater amyloid load in the orbital frontal, superior frontal, parietal, posterior precuneus, posterior cingulate, lateral temporal, inferior temporal and fusiform regions, and tau burden in the posterior cingulate, entorhinal and parahippocampal gyrus. Long-term donepezil treatment was also associated with worse performance on the 13-item Alzheimer's Disease Assessment Scale-Cognitive subscale after adjusting for AD-related risk factors and regional brain amyloid or tau load. These results indicate that long-term donepezil treatment is associated with increased regional amyloid and tau burden and worse cognitive performance among individuals with MCI. Our study highlights the importance of using noninvasive and quantitative 18 F-AV-45 and 18 F-AV-1451 PET to elucidate the consequences of drug administration in AD studies.
Collapse
Affiliation(s)
- Jian Li
- Department of Nuclear Medicine (PET Center), Xiangya Hospital Central South University, Changsha, China.,Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chaojie Zheng
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA.,Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China
| | - Qi Ge
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China
| | - Shaozhen Yan
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Manish D Paranjpe
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
| | - Shuo Hu
- Department of Nuclear Medicine (PET Center), Xiangya Hospital Central South University, Changsha, China.,Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, China
| | - Yun Zhou
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA.,Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China
| | | |
Collapse
|
20
|
Paranjpe MD, Belonwu S, Wang JK, Oskotsky T, Gupta A, Taubes A, Zalocusky KA, Paranjpe I, Glicksberg BS, Huang Y, Sirota M. Sex-Specific Cross Tissue Meta-Analysis Identifies Immune Dysregulation in Women With Alzheimer's Disease. Front Aging Neurosci 2021; 13:735611. [PMID: 34658838 PMCID: PMC8515049 DOI: 10.3389/fnagi.2021.735611] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/01/2021] [Indexed: 01/09/2023] Open
Abstract
Background: Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common cause of dementia in the United States. In spite of evidence of females having a greater lifetime risk of developing Alzheimer's Disease (AD) and greater apolipoprotein E4-related (APOE ε4) AD risk compared to males, molecular signatures underlying these differences remain elusive. Methods: We took a meta-analysis approach to study gene expression in the brains of 1,084 AD patients and age-matched controls and whole blood from 645 AD patients and age-matched controls in seven independent datasets. Sex-specific gene expression patterns were investigated through use of gene-based, pathway-based and network-based approaches. The ability of a sex-specific AD gene expression signature to distinguish Alzheimer's disease from healthy controls was assessed using a linear support vector machine model. Cell type deconvolution from whole blood gene expression data was performed to identify differentially regulated cells in males and females with AD. Results: Strikingly gene-expression, network-based analysis and cell type deconvolution approaches revealed a consistent immune signature in the brain and blood of female AD patients that was absent in males. In females, network-based analysis revealed a coordinated program of gene expression involving several zinc finger nuclease genes related to Herpes simplex viral infection whose expression was modulated by the presence of the APOE ε4 allele. Interestingly, this gene expression program was missing in the brains of male AD patients. Cell type deconvolution identified an increase in neutrophils and naïve B cells and a decrease in M2 macrophages, memory B cells, and CD8+ T cells in AD samples compared to controls in females. Interestingly, among males with AD, no significant differences in immune cell proportions compared to controls were observed. Machine learning-based classification of AD using gene expression from whole blood in addition to clinical features produced an improvement in classification accuracy upon stratifying by sex, achieving an AUROC of 0.91 for females and 0.80 for males. Conclusion: These results help identify sex and APOE ε4 genotype-specific transcriptomic signatures of AD and underscore the importance of considering sex in the development of biomarkers and therapeutic strategies for AD.
Collapse
Affiliation(s)
- Manish D Paranjpe
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States.,Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, United States
| | - Stella Belonwu
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States.,Pharmaceutical Sciences and Pharmacogenomics Graduate Program, University of California, San Francisco, San Francisco, CA, United States
| | - Jason K Wang
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, United States
| | - Tomiko Oskotsky
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Aarzu Gupta
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Alice Taubes
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States.,The Gladstone Institute of Neurological Disease, San Francisco, CA, United States
| | - Kelly A Zalocusky
- The Gladstone Institute of Neurological Disease, San Francisco, CA, United States.,Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Ishan Paranjpe
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States.,Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Benjamin S Glicksberg
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Yadong Huang
- The Gladstone Institute of Neurological Disease, San Francisco, CA, United States.,Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States.,Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
21
|
Farmer BC, Williams HC, Devanney NA, Piron MA, Nation GK, Carter DJ, Walsh AE, Khanal R, Young LEA, Kluemper JC, Hernandez G, Allenger EJ, Mooney R, Golden LR, Smith CT, Brandon JA, Gupta VA, Kern PA, Gentry MS, Morganti JM, Sun RC, Johnson LA. APOΕ4 lowers energy expenditure in females and impairs glucose oxidation by increasing flux through aerobic glycolysis. Mol Neurodegener 2021; 16:62. [PMID: 34488832 PMCID: PMC8420022 DOI: 10.1186/s13024-021-00483-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 08/15/2021] [Indexed: 01/21/2023] Open
Abstract
Background Cerebral glucose hypometabolism is consistently observed in individuals with Alzheimer’s disease (AD), as well as in young cognitively normal carriers of the Ε4 allele of Apolipoprotein E (APOE), the strongest genetic predictor of late-onset AD. While this clinical feature has been described for over two decades, the mechanism underlying these changes in cerebral glucose metabolism remains a critical knowledge gap in the field. Methods Here, we undertook a multi-omic approach by combining single-cell RNA sequencing (scRNAseq) and stable isotope resolved metabolomics (SIRM) to define a metabolic rewiring across astrocytes, brain tissue, mice, and human subjects expressing APOE4. Results Single-cell analysis of brain tissue from mice expressing human APOE revealed E4-associated decreases in genes related to oxidative phosphorylation, particularly in astrocytes. This shift was confirmed on a metabolic level with isotopic tracing of 13C-glucose in E4 mice and astrocytes, which showed decreased pyruvate entry into the TCA cycle and increased lactate synthesis. Metabolic phenotyping of E4 astrocytes showed elevated glycolytic activity, decreased oxygen consumption, blunted oxidative flexibility, and a lower rate of glucose oxidation in the presence of lactate. Together, these cellular findings suggest an E4-associated increase in aerobic glycolysis (i.e. the Warburg effect). To test whether this phenomenon translated to APOE4 humans, we analyzed the plasma metabolome of young and middle-aged human participants with and without the Ε4 allele, and used indirect calorimetry to measure whole body oxygen consumption and energy expenditure. In line with data from E4-expressing female mice, a subgroup analysis revealed that young female E4 carriers showed a striking decrease in energy expenditure compared to non-carriers. This decrease in energy expenditure was primarily driven by a lower rate of oxygen consumption, and was exaggerated following a dietary glucose challenge. Further, the stunted oxygen consumption was accompanied by markedly increased lactate in the plasma of E4 carriers, and a pathway analysis of the plasma metabolome suggested an increase in aerobic glycolysis. Conclusions Together, these results suggest astrocyte, brain and system-level metabolic reprogramming in the presence of APOE4, a ‘Warburg like’ endophenotype that is observable in young females decades prior to clinically manifest AD. Supplementary Information The online version contains supplementary material available at 10.1186/s13024-021-00483-y.
Collapse
Affiliation(s)
- Brandon C Farmer
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Holden C Williams
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA.,Sanders Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Nicholas A Devanney
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA.,Sanders Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Margaret A Piron
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Grant K Nation
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - David J Carter
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Adeline E Walsh
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Rebika Khanal
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Lyndsay E A Young
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Jude C Kluemper
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Gabriela Hernandez
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Elizabeth J Allenger
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Rachel Mooney
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Lesley R Golden
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Cathryn T Smith
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - J Anthony Brandon
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Vedant A Gupta
- Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY, USA
| | - Philip A Kern
- Center for Clinical and Translational Science, University of Kentucky College of Medicine, Lexington, KY, USA.,Department of Internal Medicine, Division of Endocrinology, University of Kentucky, Lexington, KY, USA
| | - Matthew S Gentry
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Josh M Morganti
- Sanders Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, USA.,Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Ramon C Sun
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Lance A Johnson
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA. .,Sanders Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, USA.
| |
Collapse
|
22
|
Zhou Y, Flores S, Mansor S, Hornbeck RC, Tu Z, Perlmutter JS, Ances B, Morris JC, Gropler RJ, Benzinger TLS. Spatially constrained kinetic modeling with dual reference tissues improves 18F-flortaucipir PET in studies of Alzheimer disease. Eur J Nucl Med Mol Imaging 2021; 48:3172-3186. [PMID: 33599811 PMCID: PMC8371062 DOI: 10.1007/s00259-020-05134-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 11/20/2020] [Indexed: 01/03/2023]
Abstract
PURPOSE Recent studies have shown that standard compartmental models using plasma input or the cerebellum reference tissue input are generally not reliable for quantifying tau burden in dynamic 18F-flortaucipir PET studies of Alzheimer disease. So far, the optimal reference region for estimating 18F-flortaucipir delivery and specific tau binding has yet to be determined. The objective of the study is to improve 18F-flortaucipir brain tau PET quantification using a spatially constrained kinetic model with dual reference tissues. METHODS Participants were classified as either cognitively normal (CN) or cognitively impaired (CI) based on clinical assessment. T1-weighted structural MRI and 105-min dynamic 18F-flortaucipir PET scans were acquired for each participant. Using both a simplified reference tissue model (SRTM2) and Logan plot with either cerebellum gray matter or centrum semiovale (CS) white matter as the reference tissue, we estimated distribution volume ratios (DVRs) and the relative transport rate constant R1 for region of interest-based (ROI) and voxelwise-based analyses. Conventional linear regression (LR) and LR with spatially constrained (LRSC) parametric imaging algorithms were then evaluated. Noise-induced bias in the parametric images was compared to estimates from ROI time activity curve-based kinetic modeling. We finally evaluated standardized uptake value ratios at early phase (SUVREP, 0.7-2.9 min) and late phase (SUVRLP, 80-105 min) to approximate R1 and DVR, respectively. RESULTS The percent coefficients of variation of R1 and DVR estimates from SRTM2 with spatially constrained modeling were comparable to those from the Logan plot and SUVRs. The SRTM2 using CS reference tissue with LRSC reduced noise-induced underestimation in the LR generated DVR images to negligible levels (< 1%). Inconsistent overestimation of DVR in the SUVRLP only occurred using the cerebellum reference tissue-based measurements. The CS reference tissue-based DVR and SUVRLP, and cerebellum-based SUVREP and R1 provided higher Cohen's effect size d to detect increased tau deposition and reduced relative tracer transport rate in CI individuals. CONCLUSION Using a spatially constrained kinetic model with dual reference tissues significantly improved quantification of relative perfusion and tau binding. Cerebellum and CS are the suggested reference tissues to estimate R1 and DVR, respectively, for dynamic 18F-flortaucipir PET studies. Cerebellum-based SUVREP and CS-based SUVRLP may be used to simplify 18F-flortaucipir PET study.
Collapse
Affiliation(s)
- Yun Zhou
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Campus Box 8225, 510 S. Kingshighway Blvd, St Louis, MO, 63110, USA.
| | - Shaney Flores
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Campus Box 8225, 510 S. Kingshighway Blvd, St Louis, MO, 63110, USA
| | - Syahir Mansor
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Campus Box 8225, 510 S. Kingshighway Blvd, St Louis, MO, 63110, USA
| | - Russ C Hornbeck
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Campus Box 8225, 510 S. Kingshighway Blvd, St Louis, MO, 63110, USA
| | - Zhude Tu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Campus Box 8225, 510 S. Kingshighway Blvd, St Louis, MO, 63110, USA
| | - Joel S Perlmutter
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Campus Box 8225, 510 S. Kingshighway Blvd, St Louis, MO, 63110, USA
- Departments of Neurology and Neuroscience, Programs of Physical Therapy and Occupational Therapy, Washington University School of Medicine, Saint Louis, MO, USA
| | - Beau Ances
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - John C Morris
- Departments of Neurology and Neuroscience, Programs of Physical Therapy and Occupational Therapy, Washington University School of Medicine, Saint Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Robert J Gropler
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Campus Box 8225, 510 S. Kingshighway Blvd, St Louis, MO, 63110, USA
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Campus Box 8225, 510 S. Kingshighway Blvd, St Louis, MO, 63110, USA
- Departments of Neurology and Neuroscience, Programs of Physical Therapy and Occupational Therapy, Washington University School of Medicine, Saint Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
23
|
Koutsodendris N, Nelson MR, Rao A, Huang Y. Apolipoprotein E and Alzheimer's Disease: Findings, Hypotheses, and Potential Mechanisms. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2021; 17:73-99. [PMID: 34460318 DOI: 10.1146/annurev-pathmechdis-030421-112756] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder that involves dysregulation of many cellular and molecular processes. It is notoriously difficult to develop therapeutics for AD due to its complex nature. Nevertheless, recent advancements in imaging technology and the development of innovative experimental techniques have allowed researchers to perform in-depth analyses to uncover the pathogenic mechanisms of AD. An important consideration when studying late-onset AD is its major genetic risk factor, apolipoprotein E4 (apoE4). Although the exact mechanisms underlying apoE4 effects on AD initiation and progression are not fully understood, recent studies have revealed critical insights into the apoE4-induced deficits that occur in AD. In this review, we highlight notable studies that detail apoE4 effects on prominent AD pathologies, including amyloid-β, tau pathology, neuroinflammation, and neural network dysfunction. We also discuss evidence that defines the physiological functions of apoE and outlines how these functions are disrupted in apoE4-related AD. Expected final online publication date for the Annual Review of Pathology: Mechanisms of Disease, Volume 17 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Nicole Koutsodendris
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, California 94131, USA; , .,Gladstone Institutes of Neurological Disease, San Francisco, California 94158, USA
| | - Maxine R Nelson
- Gladstone Institutes of Neurological Disease, San Francisco, California 94158, USA.,Biomedical Sciences Graduate Program, University of California, San Francisco, California 94143, USA
| | - Antara Rao
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, California 94131, USA; , .,Gladstone Institutes of Neurological Disease, San Francisco, California 94158, USA
| | - Yadong Huang
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, California 94131, USA; , .,Gladstone Institutes of Neurological Disease, San Francisco, California 94158, USA.,Biomedical Sciences Graduate Program, University of California, San Francisco, California 94143, USA.,Department of Neurology, University of California, San Francisco, California 94158, USA
| |
Collapse
|
24
|
Longitudinal analysis of APOE-ε4 genotype with the logical memory delayed recall score in Alzheimer’s disease. J Genet 2021. [DOI: 10.1007/s12041-021-01309-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
25
|
Yoon HJ, Kim BS, Jeong JH, Kim GH, Park HK, Chun MY, Ha S. Dual-phase 18F-florbetaben PET provides cerebral perfusion proxy along with beta-amyloid burden in Alzheimer's disease. NEUROIMAGE-CLINICAL 2021; 31:102773. [PMID: 34339946 PMCID: PMC8346681 DOI: 10.1016/j.nicl.2021.102773] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 07/13/2021] [Accepted: 07/20/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND This study investigated changes in brain perfusion and Aβ burden according to the progression of Alzheimer's disease (AD) by using a dual-phase 18F-florbetaben (FBB) PET protocol. METHODS Sixty subjects, including 12 with Aβ-negative normal cognition (Aβ-NC), 32 with Aβ-positive mild cognitive impairment (Aβ+MCI), and 16 with Aβ-positive AD (Aβ+AD), were enrolled. A dynamic PET scan was obtained in the early phase (0-10 min, eFBB) and delayed phase (90-110 min, dFBB), which were then averaged into a single frame, respectively. In addition to the averaged eFBB, an R1 parametric map was calculated from the eFBB scan based on a simplified reference tissue model (SRTM). Between-group regional and voxel-wise analyses of the images were performed. The associations between cognitive profiles and PET-derived parameters were investigated. RESULTS Both the R1 and eFBB perfusion reductions in the cortical regions were not significantly different between the Aβ-NC and Aβ+MCI groups, while they were significantly reduced from the Aβ+MCI to Aβ+AD groups in regional and voxel-wise analyses. However, cortical Aβ depositions on dFBB were not significantly different between the Aβ+MCI and Aβ+AD groups. There were strong positive correlations between the R1 and eFBB images in regional and voxel-wise analyses. Both perfusion components showed significant correlations with general and specific cognitive profiles. CONCLUSION The results of this study demonstrated the feasibility of dual-phase 18F-FBB PET to evaluate different trajectories of dual biomarkers for neurodegeneration and Aβ burden over the course of AD. In addition, both eFBB and SRTM-based R1 can provide robust indices of brain perfusion.
Collapse
Affiliation(s)
- Hai-Jeon Yoon
- Department of Nuclear Medicine, Ewha Womans University, School of Medicine, Seoul, Republic of Korea
| | - Bom Sahn Kim
- Department of Nuclear Medicine, Ewha Womans University, School of Medicine, Seoul, Republic of Korea.
| | - Jee Hyang Jeong
- Department of Neurology, Ewha Womans University School of Medicine, Republic of Korea.
| | - Geon Ha Kim
- Department of Neurology, Ewha Womans University School of Medicine, Republic of Korea
| | - Hee Kyung Park
- Department of Neurology, Ewha Womans University School of Medicine, Republic of Korea; Division of Psychiatry, Department of mental health care of older people, University College London, London, UK
| | - Min Young Chun
- Department of Neurology, Ewha Womans University School of Medicine, Republic of Korea
| | - Seunggyun Ha
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
26
|
Yan S, Zheng C, Paranjpe MD, Li Y, Li W, Wang X, Benzinger TLS, Lu J, Zhou Y. Sex modifies APOE ε4 dose effect on brain tau deposition in cognitively impaired individuals. Brain 2021; 144:3201-3211. [PMID: 33876815 PMCID: PMC8634082 DOI: 10.1093/brain/awab160] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/16/2021] [Accepted: 04/08/2021] [Indexed: 11/23/2022] Open
Abstract
Recent studies in cognitively unimpaired elderly individuals suggest that the APOE ε4 allele exerts a dosage-dependent effect on brain tau deposition. The aim of this study was to investigate sex differences in APOE ε4 gene dosage effects on brain tau deposition in cognitively impaired individuals using quantitative 18F-flortaucipir PET. Preprocessed 18F-flortaucipir tau PET images, T1-weighted structural MRI, demographic information, global cortical amyloid-β burden measured by 18F-florbetapir PET, CSF total tau and phosphorylated tau measurements were obtained from the Alzheimer’s Disease Neuroimaging Initiative database. Two hundred and sixty-eight cognitively impaired individuals with 146 APOE ε4 non-carriers and 122 carriers (85 heterozygotes and 37 homozygotes) were included in the study. An iterative reblurred Van Cittert iteration partial volume correction method was applied to all downloaded PET images. Magnetic resonance images were used for PET spatial normalization. Twelve regional standardized uptake value ratios relative to the cerebellum were computed in standard space. APOE ε4 dosage × sex interaction effect on 18F-flortaucipir standardized uptake value ratios was assessed using generalized linear models and sex-stratified analysis. We observed a significant APOE ε4 dosage × sex interaction effect on tau deposition in the lateral temporal, posterior cingulate, medial temporal, inferior temporal, entorhinal cortex, amygdala, parahippocampal gyrus regions after adjusting for age and education level (P < 0.05). The medial temporal, entorhinal cortex, amygdala and parahippocampal gyrus regions retained a significant APOE ε4 dosage × sex interaction effect on tau deposition after adjusting for global cortical amyloid-β (P < 0.05). In sex-stratified analysis, there was no significant difference in tau deposition between female homozygotes and heterozygotes (P > 0.05). In contrast, male homozygotes standardized uptake value ratios were significantly greater than heterozygotes or non-carriers throughout all 12 regions of interest (P < 0.05). Female heterozygotes exhibited significantly increased tau deposition compared to male heterozygotes in the orbitofrontal, posterior cingulate, lateral temporal, inferior temporal, entorhinal cortex, amygdala and parahippocampal gyrus (P < 0.05). Results from voxel-wise analysis were similar to the ones obtained from regions of interest analysis. Our findings indicate that an APOE ε4 dosage effect on brain region-specific tau deposition exists in males, but not females. These results have important clinical implications towards developing sex and genotype-guided therapeutics in Alzheimer’s disease and uncovers a potential explanation underlying differential APOE ε4-associated Alzheimer’s risk in males and females.
Collapse
Affiliation(s)
- Shaozhen Yan
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Chaojie Zheng
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.,Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China
| | - Manish D Paranjpe
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
| | - Yanxiao Li
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China.,School of Computer Science, the University of Sydney, NSW 2006, Australia
| | - Weihua Li
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiuying Wang
- School of Computer Science, the University of Sydney, NSW 2006, Australia
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.,Department of Neurology, Washington University in St. Louis School of Medicine, Saint Louis, MO, USA
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yun Zhou
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.,Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China
| | | |
Collapse
|
27
|
Mole JP, Fasano F, Evans J, Sims R, Kidd E, Aggleton JP, Metzler-Baddeley C. APOE-ε4-related differences in left thalamic microstructure in cognitively healthy adults. Sci Rep 2020; 10:19787. [PMID: 33188215 PMCID: PMC7666117 DOI: 10.1038/s41598-020-75992-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/15/2020] [Indexed: 01/05/2023] Open
Abstract
APOE-ε4 is a main genetic risk factor for developing late onset Alzheimer's disease (LOAD) and is thought to interact adversely with other risk factors on the brain. However, evidence regarding the impact of APOE-ε4 on grey matter structure in asymptomatic individuals remains mixed. Much attention has been devoted to characterising APOE-ε4-related changes in the hippocampus, but LOAD pathology is known to spread through the whole of the Papez circuit including the limbic thalamus. Here, we tested the impact of APOE-ε4 and two other risk factors, a family history of dementia and obesity, on grey matter macro- and microstructure across the whole brain in 165 asymptomatic individuals (38-71 years). Microstructural properties of apparent neurite density and dispersion, free water, myelin and cell metabolism were assessed with Neurite Orientation Density and Dispersion (NODDI) and quantitative magnetization transfer (qMT) imaging. APOE-ε4 carriers relative to non-carriers had a lower macromolecular proton fraction (MPF) in the left thalamus. No risk effects were present for cortical thickness, subcortical volume, or NODDI indices. Reduced thalamic MPF may reflect inflammation-related tissue swelling and/or myelin loss in APOE-ε4. Future prospective studies should investigate the sensitivity and specificity of qMT-based MPF as a non-invasive biomarker for LOAD risk.
Collapse
Affiliation(s)
- Jilu P Mole
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Fabrizio Fasano
- Siemens Healthcare, Henkestrasse 127, 91052, Erlangen, Germany
| | - John Evans
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Rebecca Sims
- Division of Psychological Medicine and Clinical Neuroscience, School of Medicine, Cardiff University, Haydn Ellis Building, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Emma Kidd
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue,, Cardiff, CF10 3NB, UK
| | - John P Aggleton
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Claudia Metzler-Baddeley
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK.
| |
Collapse
|
28
|
Yan S, Zheng C, Paranjpe MD, Li J, Benzinger TL, Lu J, Zhou Y. Association of sex and APOE ε4 with brain tau deposition and atrophy in older adults with Alzheimer's disease. Theranostics 2020; 10:10563-10572. [PMID: 32929366 PMCID: PMC7482805 DOI: 10.7150/thno.48522] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/04/2020] [Indexed: 11/05/2022] Open
Abstract
The objective of this study was to assess the association of sex and the apolipoprotein E (APOE) ε4 allele with brain tau deposition and atrophy in older adults with Alzheimer's disease (AD) using quantitative 18F-AV-1451 positron emission tomography (PET) and magnetic resonance imaging (MRI). Methods: Preprocessed 18F-AV-1451 tau PET, raw T1-weighted structural MR images, demographic information, cerebrospinal fluid (CSF) total tau (t-tau) and phosphorylated tau (p-tau) measurements from 57 elderly individuals with AD were downloaded from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database. An iteratively reblurred Van Cittert partial volume correction (PVC) method was applied to all preprocessed PET images. MRI images were used for PET spatial normalization and gray matter volume calculation. 18F-AV-1451 PET standardized uptake value ratio (SUVR) was calculated relative to the cerebellum gray matter. The effect of sex and APOE ε4 status on SUVR and gray matter volume were assessed at both region of interest (ROI) and voxelwise levels. Results: Female APOE ε4 carriers (FACs) had significant higher 18F-AV-1451 SUVRs in the lateral temporal, parietal, posterior cingulate, medial temporal, inferior temporal, entorhinal cortex, amygdala and parahippocampal gyrus regions, and exhibited smaller gray matter volumes in the posterior cingulate, medial temporal, inferior temporal and amygdala regions, as compared to the non-FACs (NFACs) comprised of female APOE ε4 non-carriers, male APOE ε4 carriers and male APOE ε4 non-carriers. Voxelwise analysis revealed forebrain and limbic clusters with greater 18F-AV-1451 SUVRs and lower gray matter volume between FACs compared to the NFACs. Negative correlations between ROI 18F-AV-1451 SUVRs and gray matter volumes were significant after adjusting for age and years of education. Conclusions: Among elderly individuals with AD, sex modified the effects of the APOE ε4 allele on region-specific tau deposition and gray matter volume. FACs had elevated brain region-specific tau PET SUVR and decreased gray matter volume in comparison to NFACs. The study provides a basis for the use of precision medicine in the diagnosis of AD and evaluation of therapeutics using 18F-AV-1451 PET and structural MRI.
Collapse
Affiliation(s)
- Shaozhen Yan
- Department of Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Chaojie Zheng
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Manish D Paranjpe
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
| | - Jian Li
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Tammie L.S. Benzinger
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington in St. Louis University School of Medicine, St. Louis, MO, USA
| | - Jie Lu
- Department of Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yun Zhou
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
29
|
Wang W, Zhao F, Ma X, Perry G, Zhu X. Mitochondria dysfunction in the pathogenesis of Alzheimer's disease: recent advances. Mol Neurodegener 2020; 15:30. [PMID: 32471464 PMCID: PMC7257174 DOI: 10.1186/s13024-020-00376-6] [Citation(s) in RCA: 618] [Impact Index Per Article: 123.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 04/24/2020] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is one of the most prevalent neurodegenerative diseases, characterized by impaired cognitive function due to progressive loss of neurons in the brain. Under the microscope, neuronal accumulation of abnormal tau proteins and amyloid plaques are two pathological hallmarks in affected brain regions. Although the detailed mechanism of the pathogenesis of AD is still elusive, a large body of evidence suggests that damaged mitochondria likely play fundamental roles in the pathogenesis of AD. It is believed that a healthy pool of mitochondria not only supports neuronal activity by providing enough energy supply and other related mitochondrial functions to neurons, but also guards neurons by minimizing mitochondrial related oxidative damage. In this regard, exploration of the multitude of mitochondrial mechanisms altered in the pathogenesis of AD constitutes novel promising therapeutic targets for the disease. In this review, we will summarize recent progress that underscores the essential role of mitochondria dysfunction in the pathogenesis of AD and discuss mechanisms underlying mitochondrial dysfunction with a focus on the loss of mitochondrial structural and functional integrity in AD including mitochondrial biogenesis and dynamics, axonal transport, ER-mitochondria interaction, mitophagy and mitochondrial proteostasis.
Collapse
Affiliation(s)
- Wenzhang Wang
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106 USA
| | - Fanpeng Zhao
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106 USA
| | - Xiaopin Ma
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106 USA
| | - George Perry
- College of Sciences, University of Texas at San Antonio, San Antonio, TX USA
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106 USA
| |
Collapse
|
30
|
Beta-amyloid (Aβ) uptake by PET imaging in older HIV+ and HIV- individuals. J Neurovirol 2020; 26:382-390. [PMID: 32270469 DOI: 10.1007/s13365-020-00836-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 02/13/2020] [Accepted: 02/24/2020] [Indexed: 10/24/2022]
Abstract
The causes of cognitive impairment among older HIV+ individuals may overlap with causes among elderly HIV seronegative (HIV-) individuals. The objective of this study was to determine if beta-amyloid (Aβ) deposition measured by [18F] AV-45 (florbetapir) positron emission tomography (PET) is increased in older HIV+ individuals compared to HIV- individuals. Forty-eight HIV+ and 25 HIV- individuals underwent [18F] AV-45 PET imaging. [18F] AV-45 binding to Aβ was measured by standardized uptake value ratios (SUVR) relative to the cerebellum in 16 cortical and subcortical regions of interest. Global and regional cortical SUVRs were compared by (1) serostatus, (2) HAND stage, and (3) age decade, comparing individuals in their 50s and > 60s. There were no differences in median global cortical SUVR stratified by HIV serostatus or HAND stage. The proportion of HIV+ participants in their 50s with elevated global amyloid uptake (SUVR > 1.40) was significantly higher than the proportion in HIV- participants (67% versus 25%, p = 0.04), and selected regional SUVR values were also higher (p < 0.05) in HIV+ compared to HIV- participants in their 50s. However, these group differences were not seen in participants in their 60s. In conclusion, PET imaging found no differences in overall global Aβ deposition stratified by HIV serostatus or HAND stage. Although there was some evidence of increased Aβ deposition in HIV+ individuals in their 50s compared to HIV- individuals which might indicate premature aging, the most parsimonious explanation for this is the relatively small sample size in this cross-sectional cohort study.
Collapse
|
31
|
Multiparametric imaging hippocampal neurodegeneration and functional connectivity with simultaneous PET/MRI in Alzheimer's disease. Eur J Nucl Med Mol Imaging 2020; 47:2440-2452. [PMID: 32157432 PMCID: PMC7396401 DOI: 10.1007/s00259-020-04752-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/03/2020] [Indexed: 12/12/2022]
Abstract
Purpose The objective of this study is to investigate the hippocampal neurodegeneration and its associated aberrant functions in mild cognitive impairment (MCI) and Alzheimer’s disease (AD) patients using simultaneous PET/MRI. Methods Forty-two cognitively normal controls (NC), 38 MCI, and 22 AD patients were enrolled in this study. All subjects underwent 18F-FDG PET/functional MRI (fMRI) and high-resolution T1-weighted MRI scans on a hybrid GE Signa PET/MRI scanner. Neurodegeneration in hippocampus and its subregions was quantified by regional gray matter volume and 18F-FDG standardized uptake value ratio (SUVR) relative to cerebellum. An iterative reblurred Van Cittert iteration method was used for voxelwise partial volume correction on 18F-FDG PET images. Regional gray matter volume was estimated from voxel-based morphometric analysis with MRI. fMRI data were analyzed after slice time correction and head motion correction using statistical parametric mapping (SPM12) with DPARSF toolbox. The regions of interest including hippocampus, cornu ammonis (CA1), CA2/3/dentate gyrus (DG), and subiculum were defined in the standard MNI space. Results Patient groups had reduced SUVR, gray matter volume, and functional connectivity compared to NC in CA1, CA2/3/DG, and subiculum (AD < MCI < NC). There was a linear correlation between the left CA2/3DG gray matter volume and 18F-FDG SUVR in AD patients (P < 0.001, r = 0.737). Significant correlation was also found between left CA2/3/DG-superior medial frontal gyrus functional connectivity and left CA2/3/DG hypometabolism in patients with AD. The functional connectivity of right CA1-precuneus in patients with MCI and right subiculum-superior frontal gyrus in patients with AD was positively correlated with mini mental status examination scores (P < 0.05). Conclusion Our findings demonstrate that the associations existed at subregional hippocampal level between the functional connectivity measured by fMRI and neurodegeneration measured by structural MRI and 18F-FDG PET. Our results may provide a basis for precision neuroimaging of hippocampus in AD.
Collapse
|
32
|
Paranjpe MD, Taubes A, Sirota M. Insights into Computational Drug Repurposing for Neurodegenerative Disease. Trends Pharmacol Sci 2019; 40:565-576. [PMID: 31326236 PMCID: PMC6771436 DOI: 10.1016/j.tips.2019.06.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/26/2019] [Accepted: 06/12/2019] [Indexed: 12/14/2022]
Abstract
Computational drug repurposing has the ability to remarkably reduce drug development time and cost in an era where these factors are prohibitively high. Several examples of successful repurposed drugs exist in fields such as oncology, diabetes, leprosy, inflammatory bowel disease, among others, however computational drug repurposing in neurodegenerative disease has presented several unique challenges stemming from the lack of validation methods and difficulty in studying heterogenous diseases of aging. Here, we examine existing approaches to computational drug repurposing, including molecular, clinical, and biophysical methods, and propose data sources and methods to advance computational drug repurposing in neurodegenerative disease using Alzheimer's disease as an example.
Collapse
Affiliation(s)
- Manish D Paranjpe
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA 94158, USA.
| | - Alice Taubes
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA 94158, USA; Gladstone Institutes, San Francisco, CA 94158, USA.
| |
Collapse
|
33
|
Liu M, Paranjpe MD, Zhou X, Duy PQ, Goyal MS, Benzinger TL, Lu J, Wang R, Zhou Y. Sex modulates the ApoE ε4 effect on brain tau deposition measured by 18F-AV-1451 PET in individuals with mild cognitive impairment. Theranostics 2019; 9:4959-4970. [PMID: 31410194 PMCID: PMC6691387 DOI: 10.7150/thno.35366] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 05/20/2019] [Indexed: 01/25/2023] Open
Abstract
The strongest genetic risk factor for Alzheimer's disease (AD) is the Apolipoprotein E type 4 allele (ApoE ε4). The interaction between sex and ApoE ε4 carrier status on AD risk remains an area of intense investigation. We hypothesized that sex modulates the relationship between ApoE ε4 carrier status and brain tau deposition (a quantitative endophenotype in AD) in individuals with mild cognitive impairment (MCI). Methods: Preprocessed 18F-AV-1451 tau and 18F-AV-45 amyloid PET images, T1-weighted structural magnetic resonance imaging (MRI) scans, demographic information, and cerebrospinal fluid (CSF) total tau (t-tau) and phosphorylated tau (p-tau) measurements from 108 MCI subjects in the Alzheimer's Disease Neuroimaging Initiative (ADNI) database were included. After downloading pre-processed images from ADNI, an iterative reblurred Van Cittertiteration partial volume correction (PVC) method was applied to all PET images. MRIs were used for PET spatial normalization. Regions of interest (ROIs) were defined in standard space, and standardized uptake value ratio (SUVR) images relative to cerebellum were computed. ApoE ε4 by sex interaction analyses on 18F-AV-1451 and CSF tau (t-tau, p-tau) were assessed using generalized linear models. The association between 18F-AV-1451 SUVR and CSF tau (t-tau, p-tau) was assessed. Results: After applying PVC and controlling for age, education level and global cortical 18F-AV-45 SUVR, we found that the entorhinal cortex, amygdala, parahippocampal gyrus, posterior cingulate, and occipital ROIs exhibited a significant ApoE ε4 by sex interaction effect (false discovery rate P < 0.1) among MCI individuals. We also found a significant ApoE ε4 by sex interaction effect on CSF t-tau and p-tau. 18F-AV-1451 SUVR in the 5 ROIs with ApoE ε4 by sex interaction was significantly correlated with CSF p-tau and t-tau. Conclusions: Our findings suggest that women are more susceptible to ApoE ε4-associated accumulation of neurofibrillary tangles in MCI compared to males. Both CSF tau (p-tau, t-tau) and brain tau PET are robust quantitative biomarkers for studying ApoE ε4 by sex effects on brain tau deposition in MCI participants.
Collapse
|
34
|
Zhao Q, Liu M, Ha L, Zhou Y. Quantitative 18F-AV1451 Brain Tau PET Imaging in Cognitively Normal Older Adults, Mild Cognitive Impairment, and Alzheimer's Disease Patients. Front Neurol 2019; 10:486. [PMID: 31156534 PMCID: PMC6530456 DOI: 10.3389/fneur.2019.00486] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 04/23/2019] [Indexed: 01/09/2023] Open
Abstract
Recent developments of tau Positron Emission Tomography (PET) allows assessment of regional neurofibrillary tangles (NFTs) deposition in human brain. Among the tau PET molecular probes, 18F-AV1451 is characterized by high selectivity for pathologic tau aggregates over amyloid plaques, limited non-specific binding in white and gray matter, and confined off-target binding. The objectives of the study are (1) to quantitatively characterize regional brain tau deposition measured by 18F-AV1451 PET in cognitively normal older adults (CN), mild cognitive impairment (MCI), and AD participants; (2) to evaluate the correlations between cerebrospinal fluid (CSF) biomarkers or Mini-Mental State Examination (MMSE) and 18F-AV1451 PET standardized uptake value ratio (SUVR); and (3) to evaluate the partial volume effects on 18F-AV1451 brain uptake. Methods: The study included total 115 participants (CN = 49, MCI = 58, and AD = 8) from the Alzheimer's Disease Neuroimaging Initiative (ADNI). Preprocessed 18F-AV1451 PET images, structural MRIs, and demographic and clinical assessments were downloaded from the ADNI database. A reblurred Van Cittertiteration method was used for voxelwise partial volume correction (PVC) on PET images. Structural MRIs were used for PET spatial normalization and region of interest (ROI) definition in standard space. The parametric images of 18F-AV1451 SUVR relative to cerebellum were calculated. The ROI SUVR measurements from PVC and non-PVC SUVR images were compared. The correlation between ROI 18F-AV1451 SUVR and the measurements of MMSE, CSF total tau (t-tau), and phosphorylated tau (p-tau) were also assessed. Results:18F-AV1451 prominently specific binding was found in the amygdala, entorhinal cortex, parahippocampus, fusiform, posterior cingulate, temporal, parietal, and frontal brain regions. Most regional SUVRs showed significantly higher uptake of 18F-AV1451 in AD than MCI and CN participants. SUVRs of small regions like amygdala, entorhinal cortex and parahippocampus were statistically improved by PVC in all groups (p < 0.01). Although there was an increasing tendency of 18F-AV-1451 SUVRs in MCI group compared with CN group, no significant difference of 18F-AV1451 deposition was found between CN and MCI brains with or without PVC (p > 0.05). Declined MMSE score was observed with increasing 18F-AV1451 binding in amygdala, entorhinal cortex, parahippocampus, and fusiform. CSF p-tau was positively correlated with 18F-AV1451 deposition. PVC improved the results of 18F-AV-1451 tau deposition and correlation studies in small brain regions. Conclusion: The typical deposition of 18F-AV1451 tau PET imaging in AD brain was found in amygdala, entorhinal cortex, fusiform and parahippocampus, and these regions were strongly associated with cognitive impairment and CSF biomarkers. Although more deposition was observed in MCI group, the 18F-AV-1451 PET imaging could not differentiate the MCI patients from CN population. More tau deposition related to decreased MMSE score and increased level of CSF p-tau, especially in ROIs of amygdala, entorhinal cortex and parahippocampus. PVC did improve the results of tau deposition and correlation studies in small brain regions and suggest to be routinely used in 18F-AV1451 tau PET quantification.
Collapse
Affiliation(s)
- Qian Zhao
- Department of Nuclear Medicine, General Hospital of Ningxia Medical University, Yinchuan, China.,The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Min Liu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Radiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Lingxia Ha
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Center for Reproductive Medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yun Zhou
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | | |
Collapse
|