1
|
Saiyisan A, Zeng S, Zhang H, Wang Z, Wang J, Cai P, Huang J. Chemical exchange saturation transfer MRI for neurodegenerative diseases: An update on clinical and preclinical studies. Neural Regen Res 2026; 21:553-568. [PMID: 39885672 DOI: 10.4103/nrr.nrr-d-24-01246] [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: 10/15/2024] [Accepted: 01/09/2025] [Indexed: 02/01/2025] Open
Abstract
Chemical exchange saturation transfer magnetic resonance imaging is an advanced imaging technique that enables the detection of compounds at low concentrations with high sensitivity and spatial resolution and has been extensively studied for diagnosing malignancy and stroke. In recent years, the emerging exploration of chemical exchange saturation transfer magnetic resonance imaging for detecting pathological changes in neurodegenerative diseases has opened up new possibilities for early detection and repetitive scans without ionizing radiation. This review serves as an overview of chemical exchange saturation transfer magnetic resonance imaging with detailed information on contrast mechanisms and processing methods and summarizes recent developments in both clinical and preclinical studies of chemical exchange saturation transfer magnetic resonance imaging for Alzheimer's disease, Parkinson's disease, multiple sclerosis, and Huntington's disease. A comprehensive literature search was conducted using databases such as PubMed and Google Scholar, focusing on peer-reviewed articles from the past 15 years relevant to clinical and preclinical applications. The findings suggest that chemical exchange saturation transfer magnetic resonance imaging has the potential to detect molecular changes and altered metabolism, which may aid in early diagnosis and assessment of the severity of neurodegenerative diseases. Although promising results have been observed in selected clinical and preclinical trials, further validations are needed to evaluate their clinical value. When combined with other imaging modalities and advanced analytical methods, chemical exchange saturation transfer magnetic resonance imaging shows potential as an in vivo biomarker, enhancing the understanding of neuropathological mechanisms in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ahelijiang Saiyisan
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Shihao Zeng
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Huabin Zhang
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Ziyan Wang
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jiawen Wang
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Pei Cai
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jianpan Huang
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Tam Wing Fan Neuroimaging Research Laboratory, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
2
|
Johnson CN, Evans MR, Blankenship AE, John CS, Rekowski MJ, Washburn MP, Phan A, Gouvion CM, Haeri M, Swerdlow RH, Geiger PC, Morris JK. Human skeletal muscle mitochondrial pathways are impacted by a neuropathologic diagnosis of Alzheimer's disease. Neurobiol Dis 2025; 210:106916. [PMID: 40250718 DOI: 10.1016/j.nbd.2025.106916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/21/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025] Open
Abstract
Alzheimer's disease (AD) is associated with reduced lean mass and impaired skeletal muscle mitochondrial and motor function. Although primary mitochondrial defects in AD may underlie these findings, molecular alterations in AD have not been thoroughly examined in human skeletal muscle. Here, we used two human skeletal muscle types, quadriceps (n = 81) and temporalis (n = 66), to compare the proteome of individuals with a neuropathologic AD diagnosis based on AD Neuropathologic Change (ADNPC+: n = 54 temporalis, 44 quadriceps) to controls (ADNPC-: n = 27 temporalis, 22 quadriceps). We determined the effects of ADNPC status within each muscle and within apolipoprotein E4 (APOE4) carriers and APOE4 non-carriers. Pathways that support mitochondrial metabolism, including oxidative phosphorylation, were downregulated in skeletal muscle of ADNPC+ versus ADNPC- individuals. Similar mitochondrial effects were observed across muscle types and APOE4 carrier groups, but nearly four times as many proteins were altered in temporalis versus quadriceps tissue and mitochondrial effects were most pronounced in APOE4 carriers compared to APOE4 non-carriers. Of all detected oxidative phosphorylation proteins, the expression of ∼29-61 % (dependent on muscle/APOE4 carrier group) significantly correlated with AD progression, ranked by Clinical Dementia Rating and ADNPC scores. Of these, 23 proteins decreased in expression with greater AD progression in all skeletal muscle type and APOE4 carrier groups. This is the first study to assess differences in the human skeletal muscle proteome in the context of AD. Our work shows that systemic mitochondrial alterations in AD extend to skeletal muscle and these effects are amplified by APOE4 and correlate with AD progression.
Collapse
Affiliation(s)
- Chelsea N Johnson
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA.
| | - Mara R Evans
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Anneka E Blankenship
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA
| | - Casey S John
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA.
| | - Michaella J Rekowski
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Michael P Washburn
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Andy Phan
- Bruker Daltonics, Inc, Billerica, MA 01821, USA.
| | - Cynthia M Gouvion
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA.
| | - Mohammad Haeri
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA; Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Russell H Swerdlow
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA; Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Paige C Geiger
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Jill K Morris
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA; Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
3
|
Wang L, Curran GL, Zhong R, Xue Z, Veerareddy V, Thieschafer J, Min PH, Li L, Lowe VJ, Kandimalla KK. Amyloid beta peptides inhibit glucose transport at the blood-brain barrier by disrupting the insulin-AKT pathway. J Cereb Blood Flow Metab 2025:271678X251332493. [PMID: 40370301 DOI: 10.1177/0271678x251332493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Molecular mechanisms underlying disruptions in brain glucose uptake and metabolism, linked with cognitive decline in Alzheimer's disease (AD) patients, are only partially understood. This study investigated how soluble amyloid beta (sAβ) peptides affect glucose transport at the blood-brain barrier (BBB), the primary portal for glucose entry into the brain. We demonstrated that [18F]-fluorodeoxyglucose (18FDG) uptake is reduced in sAβ overproducing APP,PS1 transgenic mice compared to wild-type mice. Moreover, the influx rate of 18FDG decreased in sAβ40 or sAβ42 pre-infused mice, highlighting the inhibitory effect of sAβ peptides on glucose transport at the BBB. Consistently, the expression of GLUT1, the primary glucose transporter at the BBB, is reduced in polarized human cerebral microvascular endothelial cell (hCMEC/D3) monolayers upon exposure to sAβ peptides and in Aβ-laden cerebral vasculature in vivo. The study further examined the influence of sAβ on the insulin-AKT pathway, known to regulate glucose uptake through modulation of thioredoxin-interacting protein (TXNIP) expression. Results showed that sAβ peptides suppress AKT phosphorylation and reduce GLUT1 expression by upregulating TXNIP levels in hCMEC/D3 monolayers. Co-incubation of resveratrol with sAβ peptides reduced TXNIP expression and rectified reductions in GLUT1 expression. In summary, toxic sAβ impairs BBB glucose transport by disrupting the insulin/AKT/TXNIP axis.
Collapse
Affiliation(s)
- Lushan Wang
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Geoffry L Curran
- Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Rui Zhong
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Zheng Xue
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Vaishnavi Veerareddy
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Josslen Thieschafer
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Paul H Min
- Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Ling Li
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Karunya K Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
4
|
Bøgh N, Aastrup M, Mortensen JK, Gottrup H, Blicher JU, Borghammer P, Kristensen MH, Hansen ESS, Vaeggemose M, Laustsen C. Comparison of Deuterium Metabolic Imaging with FDG PET in Alzheimer Disease. Radiology 2025; 315:e241808. [PMID: 40197092 DOI: 10.1148/radiol.241808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Background The approval of amyloid-targeting therapies has made it increasingly important to differentiate Alzheimer disease (AD) from other causes of dementia. Dysfunctional glucose metabolism is a recognized pathophysiological element in AD that may be visualized with spectroscopic MRI of deuterated glucose and its metabolites, also known as deuterium metabolic imaging (DMI). Purpose To explore the potential of DMI as a diagnostic tool for AD. Materials and Methods In this prospective cross-sectional study, participants with newly diagnosed AD and age-matched controls were recruited from April to October 2023. DMI was performed with a 3-T system equipped with a proton/deuterium head coil following oral consumption of 75 g of deuterated glucose. Clinical fluorodeoxyglucose (FDG) PET data were acquired from patient records for comparison. The predefined primary outcome, the ratio between lactate and glutamine plus glutamate (Glx) at DMI, was analyzed using age-corrected linear mixed-effect models. Results Ten participants with AD (mean age, 72 years ± 6 [SD]; six women) and five age-matched healthy controls (mean age, 68 years ± 7; four men) were included. The primary analysis revealed no evidence of a difference in the ratio of lactate to Glx between participants with AD and controls (P = .24 across all regions of interest). Exploratory analyses revealed that participants with AD had reduced signals for medial temporal lactate (0.7 ± 0.2 vs 0.5 ± 0.1, P = .04) and Glx (0.5 ± 0.03 vs 0.48 ± 0.05, P = .03) compared with controls. Finally, a strong correlation (r = 0.73) was observed between DMI and FDG PET. Conclusion This study did not find evidence to support a shift from oxidative to anaerobic metabolism in AD. Exploratory analyses revealed a decrease in glucose metabolism in the medial temporal lobe. In extension hereof, a similar distribution of low DMI metabolism and decreased FDG PET glucose uptake was observed. © RSNA, 2025 Supplemental material is available for this article. See also the article by Liu et al in this issue. See also the editorial by Port in this issue.
Collapse
Affiliation(s)
- Nikolaj Bøgh
- Department of Clinical Medicine, MR Research Centre, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Malene Aastrup
- Department of Clinical Medicine, MR Research Centre, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Janne K Mortensen
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Hanne Gottrup
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Jakob U Blicher
- Department of Neurology, Aalborg University Hospital, Aalborg, Denmark
| | - Per Borghammer
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Mattias H Kristensen
- Department of Clinical Medicine, MR Research Centre, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Esben S S Hansen
- Department of Clinical Medicine, MR Research Centre, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Michael Vaeggemose
- Department of Clinical Medicine, MR Research Centre, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
- GE HealthCare, Brondby, Denmark
| | - Christoffer Laustsen
- Department of Clinical Medicine, MR Research Centre, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| |
Collapse
|
5
|
Johnson CN, Lysaker CR, Gast EC, McCoin CS, Kemna RE, Fuller KNZ, Kugler BA, Franczak E, Csikos V, Allen J, John CS, Wolf MA, Morris ME, Thyfault JP, Wilkins HM, Geiger PC, Morris JK. APOE4 Exerts Partial Diet-dependent Effects on Energy Expenditure and Skeletal Muscle Mitochondrial Pathways in a Preclinical Model. FUNCTION 2025; 6:zqaf017. [PMID: 40133005 PMCID: PMC11980864 DOI: 10.1093/function/zqaf017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/28/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
Apolipoprotein E4 (APOE4) is the greatest genetic risk factor for Alzheimer's (AD) and is linked to whole-body metabolic dysfunction. However, it is unclear how APOE4 interacts with modifiable factors like diet to impact tissues central to regulating whole-body metabolism. We examined APOE4- and Western diet-driven effects in skeletal muscle using APOE3 (control) and APOE4 targeted replacement mice on a C57BL/6NTac background fed a high-fat diet (HFD, 45% kcal fat) or low-fat diet (LFD, 10% kcal fat) for 4 months (n = 7-8 per genotype/diet/sex combination). We assessed body composition and whole-body outcomes linked to skeletal muscle function including respiratory exchange ratio (RER) and resting energy expenditure (REE). In skeletal muscle, we evaluated the proteome and mitochondrial respiration. In males only, APOE4 drove greater gains in fat mass and lower gains in lean mass on both diets. APOE4 did not affect daily RER but was associated with elevated REE in males and lower REE in HFD females after covarying for body composition. Skeletal muscle proteomics showed APOE4 exerts several diet- and sex-specific effects on mitochondrial pathways, including elevations in branched-chain amino catabolism in HFD males and reductions in oxidative phosphorylation in LFD females. This did not translate to differences in skeletal muscle mitochondrial respiration, suggesting that compensatory mechanisms may sustain mitochondrial function at this age. Our work indicates that genetic risk may mediate early life effects on skeletal muscle mitochondria and energy expenditure that are partially dependent on diet. This has important implications for mitigating ad risk in APOE4 carriers.
Collapse
Affiliation(s)
- Chelsea N Johnson
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA
| | - Colton R Lysaker
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA
| | - Elaine C Gast
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Colin S McCoin
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- University of Kansas Diabetes Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Riley E Kemna
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA
| | - Kelly N Z Fuller
- University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Benjamin A Kugler
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Edziu Franczak
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Vivien Csikos
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA
| | - Julie Allen
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Casey S John
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA
| | - MaryJane A Wolf
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Matthew E Morris
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- University of Kansas Diabetes Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - John P Thyfault
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA
- University of Kansas Diabetes Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Heather M Wilkins
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Paige C Geiger
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- University of Kansas Diabetes Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jill K Morris
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
6
|
Kedia S, Simons M. Oligodendrocytes in Alzheimer's disease pathophysiology. Nat Neurosci 2025; 28:446-456. [PMID: 39881195 DOI: 10.1038/s41593-025-01873-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025]
Abstract
Our understanding of Alzheimer's disease (AD) has transformed from a purely neuronal perspective to one that acknowledges the involvement of glial cells. Despite remarkable progress in unraveling the biology of microglia, astrocytes and vascular elements, the exploration of oligodendrocytes in AD is still in its early stages. Contrary to the traditional notion of oligodendrocytes as passive bystanders in AD pathology, emerging evidence indicates their active participation in and reaction to amyloid and tau pathology. Oligodendrocytes undergo a functional transition to a disease-associated state, engaging in immune modulation, stress responses and cellular survival. Far from being inert players, they appear to serve a dual role in AD pathogenesis, potentially offering defense mechanisms against pathology while also contributing to disease progression. This Review explores recent advancements in understanding the roles of oligodendrocytes and their myelin sheaths in the context of AD, shedding light on their complex interactions within the disease pathology.
Collapse
Affiliation(s)
- Shreeya Kedia
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.
| |
Collapse
|
7
|
Kulminski AM, Jain‐Washburn E, Nazarian A, Wilkins HM, Veatch O, Swerdlow RH, Honea RA. Association of APOE alleles and polygenic profiles comprising APOE-TOMM40-APOC1 variants with Alzheimer's disease neuroimaging markers. Alzheimers Dement 2025; 21:e14445. [PMID: 39713891 PMCID: PMC11848341 DOI: 10.1002/alz.14445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/28/2024] [Accepted: 11/10/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION TOMM40 and APOC1 variants can modulate the APOE-ε4-related Alzheimer's disease (AD) risk by up to fourfold. We aim to investigate whether the genetic modulation of ε4-related AD risk is reflected in brain morphology. METHODS We tested whether 27 magnetic resonance imaging-derived neuroimaging markers of neurodegeneration (volume and thickness in temporo-limbic regions) are associated with APOE-TOMM40-APOC1 polygenic profiles using the National Alzheimer's Coordinating Center Uniform Data Set linked to the AD Genetic Consortium data. RESULTS All brain regions studied using structural phenotypes were smaller in individuals with AD. The ε4 allele was associated with smaller limbic (entorhinal, hippocampus, parahippocampus) brain volume and cortical thickness in AD cases than controls. There were significant differences in the associations for the higher-risk and lower-risk ε4-bearing APOE-TOMM40-APOC1 profiles with temporo-limbic region markers. DISCUSSION The APOE-AD heterogeneity may be partly attributed to the modulating role of the TOMM40 and APOC1 genes in the APOE cluster. HIGHLIGHTS The ε4 allele is associated with smaller values of neuroimaging markers in AD cases. Larger values of neuroimaging markers may protect against AD in the ε4 carriers. TOMM40 and APOC1 variants differentiate AD risk in the ε4 carriers. The same variants can differentiate the links between ε4 and neuroimaging markers.
Collapse
Affiliation(s)
- Alexander M. Kulminski
- Biodemography of Aging Research UnitSocial Science Research InstituteDuke UniversityDurhamNorth CarolinaUSA
| | - Ethan Jain‐Washburn
- Biodemography of Aging Research UnitSocial Science Research InstituteDuke UniversityDurhamNorth CarolinaUSA
| | - Alireza Nazarian
- Biodemography of Aging Research UnitSocial Science Research InstituteDuke UniversityDurhamNorth CarolinaUSA
| | - Heather M. Wilkins
- Department of NeurologyUniversity of Kansas School of MedicineKansas CityKansasUSA
- University of Kansas Alzheimer's Disease CenterUniversity of Kansas School of MedicineKansas CityKansasUSA
| | - Olivia Veatch
- University of Kansas Alzheimer's Disease CenterUniversity of Kansas School of MedicineKansas CityKansasUSA
- Department of PsychiatryUniversity of Kansas School of MedicineKansas CityKansasUSA
| | - Russell H. Swerdlow
- Department of NeurologyUniversity of Kansas School of MedicineKansas CityKansasUSA
- University of Kansas Alzheimer's Disease CenterUniversity of Kansas School of MedicineKansas CityKansasUSA
| | - Robyn A. Honea
- Department of NeurologyUniversity of Kansas School of MedicineKansas CityKansasUSA
- University of Kansas Alzheimer's Disease CenterUniversity of Kansas School of MedicineKansas CityKansasUSA
| |
Collapse
|
8
|
Roe JM, Vidal-Piñeiro D, Sørensen Ø, Grydeland H, Leonardsen EH, Iakunchykova O, Pan M, Mowinckel A, Strømstad M, Nawijn L, Milaneschi Y, Andersson M, Pudas S, Bråthen ACS, Kransberg J, Falch ES, Øverbye K, Kievit RA, Ebmeier KP, Lindenberger U, Ghisletta P, Demnitz N, Boraxbekk CJ, Drevon CA, Penninx B, Bertram L, Nyberg L, Walhovd KB, Fjell AM, Wang Y. Brain change trajectories in healthy adults correlate with Alzheimer's related genetic variation and memory decline across life. Nat Commun 2024; 15:10651. [PMID: 39690174 PMCID: PMC11652687 DOI: 10.1038/s41467-024-53548-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 10/16/2024] [Indexed: 12/19/2024] Open
Abstract
Throughout adulthood and ageing our brains undergo structural loss in an average pattern resembling faster atrophy in Alzheimer's disease (AD). Using a longitudinal adult lifespan sample (aged 30-89; 2-7 timepoints) and four polygenic scores for AD, we show that change in AD-sensitive brain features correlates with genetic AD-risk and memory decline in healthy adults. We first show genetic risk links with more brain loss than expected for age in early Braak regions, and find this extends beyond APOE genotype. Next, we run machine learning on AD-control data from the Alzheimer's Disease Neuroimaging Initiative using brain change trajectories conditioned on age, to identify AD-sensitive features and model their change in healthy adults. Genetic AD-risk linked with multivariate change across many AD-sensitive features, and we show most individuals over age ~50 are on an accelerated trajectory of brain loss in AD-sensitive regions. Finally, high genetic risk adults with elevated brain change showed more memory decline through adulthood, compared to high genetic risk adults with less brain change. Our findings suggest quantitative AD risk factors are detectable in healthy individuals, via a shared pattern of ageing- and AD-related neurodegeneration that occurs along a continuum and tracks memory decline through adulthood.
Collapse
Grants
- U01 AG024904 NIA NIH HHS
- The infrastructure for the NESDA study (www.nesda.nl) is funded through the Geestkracht program of the Netherlands Organisation for Health Research and Development (ZonMw, grant number 10-000‐1002) and financial contributions by participating universities and mental health care organizations (VU University Medical Center, GGZ inGeest, Leiden University Medical Center, Leiden University, GGZ Rivierdu-inen, University Medical Center Groningen, University of Groningen, Lentis, GGZ Friesland, GGZ Drenthe, Rob Giel Onderzoekscentrum).
- Scholar grant from Knut and Alice Wallenberg’s (KAW) foundation to L.N.
- European Research Council 313440 (to K.B.W.) Norwegian Research Council (to A.M.F. and K.B.W.) under grants 249931 (TOPPFORSK)
- European Research Council under grants 283634, 725025 (to A.M.F.) Norwegian Research Council (to A.M.F. and K.B.W.) under grants 249931 (TOPPFORSK) The National Association for Public Health’s dementia research program, Norway (to A.M.F)
- Norwegian Research Council grant 302854 (FRIPRO; to Y.W.)
Collapse
Affiliation(s)
- James M Roe
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway.
| | - Didac Vidal-Piñeiro
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
| | - Øystein Sørensen
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
| | - Håkon Grydeland
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
| | - Esten H Leonardsen
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Olena Iakunchykova
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
| | - Mengyu Pan
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Athanasia Mowinckel
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
| | - Marie Strømstad
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
| | - Laura Nawijn
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Psychiatry and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Yuri Milaneschi
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Psychiatry and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Micael Andersson
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
| | - Sara Pudas
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
| | - Anne Cecilie Sjøli Bråthen
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
| | - Jonas Kransberg
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
| | - Emilie Sogn Falch
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
| | - Knut Øverbye
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
| | - Rogier A Kievit
- Cognitive Neuroscience Department, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Klaus P Ebmeier
- Department of Psychiatry and Wellcome Centre for Integrative Neuroimaging, University of Oxford, Warneford Hospital, Oxford, United Kingdom
| | - Ulman Lindenberger
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany
- Max Planck UCL Centre for Computational Psychiatry and Ageing Research, Berlin, Germany
| | - Paolo Ghisletta
- Faculty of Psychology and Educational Sciences, University of Geneva, Geneva, Switzerland
| | - Naiara Demnitz
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| | - Carl-Johan Boraxbekk
- Institute for Clinical Medicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Radiation Sciences, Diagnostic Radiology, and Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
- Institute of Sports Medicine Copenhagen (ISMC) and Department of Neurology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Christian A Drevon
- Department of Nutrition, Institute of Basic Medical Science, Faculty of Medicine, University of Oslo, Oslo, Norway
- Vitas Ltd, Oslo Science Park, Oslo, Norway
| | - Brenda Penninx
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Psychiatry and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, Lübeck, Germany
| | - Lars Nyberg
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
- Department of Diagnostics and Intervention, Umeå University, Umeå, Sweden
- Department of Health, Education and Technology, Luleå University of Technology, Luleå, Sweden
| | - Kristine B Walhovd
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
- Computational Radiology and Artificial Intelligence, Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Anders M Fjell
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
- Computational Radiology and Artificial Intelligence, Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Yunpeng Wang
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
| |
Collapse
|
9
|
Ardanaz CG, de la Cruz A, Minhas PS, Hernández-Martín N, Pozo MÁ, Valdecantos MP, Valverde ÁM, Villa-Valverde P, Elizalde-Horcada M, Puerta E, Ramírez MJ, Ortega JE, Urbiola A, Ederra C, Ariz M, Ortiz-de-Solórzano C, Fernández-Irigoyen J, Santamaría E, Karsenty G, Brüning JC, Solas M. Astrocytic GLUT1 reduction paradoxically improves central and peripheral glucose homeostasis. SCIENCE ADVANCES 2024; 10:eadp1115. [PMID: 39423276 PMCID: PMC11488540 DOI: 10.1126/sciadv.adp1115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 09/13/2024] [Indexed: 10/21/2024]
Abstract
Astrocytes are considered an essential source of blood-borne glucose or its metabolites to neurons. Nonetheless, the necessity of the main astrocyte glucose transporter, i.e., GLUT1, for brain glucose metabolism has not been defined. Unexpectedly, we found that brain glucose metabolism was paradoxically augmented in mice with astrocytic GLUT1 reduction (GLUT1ΔGFAP mice). These mice also exhibited improved peripheral glucose metabolism especially in obesity, rendering them metabolically healthier. Mechanistically, we observed that GLUT1-deficient astrocytes exhibited increased insulin receptor-dependent ATP release, and that both astrocyte insulin signaling and brain purinergic signaling are essential for improved brain function and systemic glucose metabolism. Collectively, we demonstrate that astrocytic GLUT1 is central to the regulation of brain energetics, yet its depletion triggers a reprogramming of brain metabolism sufficient to sustain energy requirements, peripheral glucose homeostasis, and cognitive function.
Collapse
Affiliation(s)
- Carlos G. Ardanaz
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, 31008 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Aida de la Cruz
- Laboratory of Local Translation in Neurons and Glia, Achucarro Basque Centre for Neuroscience, 48940 Leioa, Spain
| | - Paras S. Minhas
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nira Hernández-Martín
- Unidad de Cartografía Cerebral, Instituto Pluridisciplinar, Universidad Complutense de Madrid, 28040 Madrid, Spain
- PET Center, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Miguel Ángel Pozo
- Unidad de Cartografía Cerebral, Instituto Pluridisciplinar, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Unidad de Cartografía Cerebral, Instituto de Investigación Sanitaria, Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - M. Pilar Valdecantos
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, Department of Metabolism and Cellular Signaling, Madrid 28029, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid 28029, Spain
- Universidad Francisco de Vitoria, Faculty of Experimental Sciences, Pozuelo de Alarcon, Madrid, Spain
| | - Ángela M. Valverde
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, Department of Metabolism and Cellular Signaling, Madrid 28029, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid 28029, Spain
| | | | | | - Elena Puerta
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, 31008 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - María J. Ramírez
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, 31008 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Jorge E. Ortega
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
- Department of Pharmacology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Biobizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Ainhoa Urbiola
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Imaging Platform, Foundation for Applied Medical Research (FIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
| | - Cristina Ederra
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Imaging Platform, Foundation for Applied Medical Research (FIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
| | - Mikel Ariz
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Imaging Platform, Foundation for Applied Medical Research (FIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
- Department of Electrical, Electronic and Communications Engineering, Public University of Navarra, 31006 Pamplona, Spain
| | - Carlos Ortiz-de-Solórzano
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Imaging Platform, Foundation for Applied Medical Research (FIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain
| | - Gerard Karsenty
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, 701 West 168th Street, New York, NY, USA
| | - Jens C. Brüning
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, 50931 Cologne, Germany
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, 50924 Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
- National Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Maite Solas
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, 31008 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| |
Collapse
|
10
|
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
|
11
|
Soldan A, Wang J, Pettigrew C, Davatzikos C, Erus G, Hohman TJ, Dumitrescu L, Bilgel M, Resnick SM, Rivera-Rivera LA, Langhough R, Johnson SC, Benzinger T, Morris JC, Laws SM, Fripp J, Masters CL, Albert MS. Alzheimer's disease genetic risk and changes in brain atrophy and white matter hyperintensities in cognitively unimpaired adults. Brain Commun 2024; 6:fcae276. [PMID: 39229494 PMCID: PMC11369827 DOI: 10.1093/braincomms/fcae276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 06/25/2024] [Accepted: 08/12/2024] [Indexed: 09/05/2024] Open
Abstract
Reduced brain volumes and more prominent white matter hyperintensities on MRI scans are commonly observed among older adults without cognitive impairment. However, it remains unclear whether rates of change in these measures among cognitively normal adults differ as a function of genetic risk for late-onset Alzheimer's disease, including APOE-ɛ4, APOE-ɛ2 and Alzheimer's disease polygenic risk scores (AD-PRS), and whether these relationships are influenced by other variables. This longitudinal study examined the trajectories of regional brain volumes and white matter hyperintensities in relationship to APOE genotypes (N = 1541) and AD-PRS (N = 1093) in a harmonized dataset of middle-aged and older individuals with normal cognition at baseline (mean baseline age = 66 years, SD = 9.6) and an average of 5.3 years of MRI follow-up (max = 24 years). Atrophy on volumetric MRI scans was quantified in three ways: (i) a composite score of regions vulnerable to Alzheimer's disease (SPARE-AD); (ii) hippocampal volume; and (iii) a composite score of regions indexing advanced non-Alzheimer's disease-related brain aging (SPARE-BA). Global white matter hyperintensity volumes were derived from fluid attenuated inversion recovery (FLAIR) MRI. Using linear mixed effects models, there was an APOE-ɛ4 gene-dose effect on atrophy in the SPARE-AD composite and hippocampus, with greatest atrophy among ɛ4/ɛ4 carriers, followed by ɛ4 heterozygouts, and lowest among ɛ3 homozygouts and ɛ2/ɛ2 and ɛ2/ɛ3 carriers, who did not differ from one another. The negative associations of APOE-ɛ4 with atrophy were reduced among those with higher education (P < 0.04) and younger baseline ages (P < 0.03). Higher AD-PRS were also associated with greater atrophy in SPARE-AD (P = 0.035) and the hippocampus (P = 0.014), independent of APOE-ɛ4 status. APOE-ɛ2 status (ɛ2/ɛ2 and ɛ2/ɛ3 combined) was not related to baseline levels or atrophy in SPARE-AD, SPARE-BA or the hippocampus, but was related to greater increases in white matter hyperintensities (P = 0.014). Additionally, there was an APOE-ɛ4 × AD-PRS interaction in relation to white matter hyperintensities (P = 0.038), with greater increases in white matter hyperintensities among APOE-ɛ4 carriers with higher AD-PRS. APOE and AD-PRS associations with MRI measures did not differ by sex. These results suggest that APOE-ɛ4 and AD-PRS independently and additively influence longitudinal declines in brain volumes sensitive to Alzheimer's disease and synergistically increase white matter hyperintensity accumulation among cognitively normal individuals. Conversely, APOE-ɛ2 primarily influences white matter hyperintensity accumulation, not brain atrophy. Results are consistent with the view that genetic factors for Alzheimer's disease influence atrophy in a regionally specific manner, likely reflecting preclinical neurodegeneration, and that Alzheimer's disease risk genes contribute to white matter hyperintensity formation.
Collapse
Affiliation(s)
- Anja Soldan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jiangxia Wang
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Corinne Pettigrew
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Christos Davatzikos
- Centre for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guray Erus
- Centre for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Timothy J Hohman
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| | - Logan Dumitrescu
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging Intramural Research Program, Baltimore, MD 21224, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging Intramural Research Program, Baltimore, MD 21224, USA
| | - Leonardo A Rivera-Rivera
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53726, USA
| | - Rebecca Langhough
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53726, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53726, USA
| | - Tammie Benzinger
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Simon M Laws
- Centre for Precision Health, Edith Cowan University, Joondalup, WA 6027, Australia
| | - Jurgen Fripp
- Australian E-Health Research Centre, CSIRO Health & Biosecurity, Herston, QLD 4029, Australia
| | - Colin L Masters
- The Florey Institute, University of Melbourne, Parkville, VIC 3052, Australia
| | - Marilyn S Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
12
|
Daly J, De Luca F, Berens SC, Field AP, Rusted JM, Bird CM. The effect of apolipoprotein E genotype on spatial processing in humans: A meta-analysis and systematic review. Cortex 2024; 177:268-284. [PMID: 38878339 DOI: 10.1016/j.cortex.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/04/2024] [Accepted: 05/20/2024] [Indexed: 07/31/2024]
Abstract
The ε4 allele of the apolipoprotein E (APOE4) gene is an established risk factor for Alzheimer's disease but its impact on cognition in healthy adults across the lifespan is unclear. One cognitive domain that is affected early in the course of Alzheimer's disease is spatial cognition, yet the evidence for APOE-related changes in spatial cognition is mixed. In this meta-analysis we assessed the impact of carrying the APOE4 allele on five subdomains of spatial cognition across the lifespan. We included studies of healthy human participants where an APOE4-carrier group (heterozygous or homozygous) could be compared to a homozygous group of APOE3-carriers. We identified 156 studies in total from three databases (Pubmed, Scopus and Web of Science) as well as through searching cited literature and contacting authors for unpublished data. 122 studies involving 32,547 participants were included in a meta-analysis, and the remaining studies are included in a descriptive review. APOE4 carriers scored significantly lower than APOE3 carriers (θˆ = -.08 [-.14, -.02]) on tests of spatial long-term memory; this effect was very small and was not modulated by age. On other subdomains of spatial cognition (spatial construction, spatial working memory, spatial reasoning, navigation) there were no effects of genotype. Overall, our results demonstrate that the APOE4 allele exerts little influence on spatial cognitive abilities in healthy adults.
Collapse
Affiliation(s)
- Jessica Daly
- Sussex Neuroscience, School of Psychology, University of Sussex, United Kingdom
| | - Flavia De Luca
- Sussex Neuroscience, School of Psychology, University of Sussex, United Kingdom
| | - Sam C Berens
- Sussex Neuroscience, School of Psychology, University of Sussex, United Kingdom
| | - Andy P Field
- School of Psychology, University of Sussex, United Kingdom
| | | | - Chris M Bird
- Sussex Neuroscience, School of Psychology, University of Sussex, United Kingdom.
| |
Collapse
|
13
|
Helgudóttir SS, Johnsen KB, Routhe LG, Rasmussen CLM, Thomsen MS, Moos T. Upregulation of Transferrin Receptor 1 (TfR1) but Not Glucose Transporter 1 (GLUT1) or CD98hc at the Blood-Brain Barrier in Response to Valproic Acid. Cells 2024; 13:1181. [PMID: 39056763 PMCID: PMC11275047 DOI: 10.3390/cells13141181] [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: 04/26/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Transferrin receptor 1 (TfR1), glucose transporter 1 (GLUT1), and CD98hc are candidates for targeted therapy at the blood-brain barrier (BBB). Our objective was to challenge the expression of TfR1, GLUT1, and CD98hc in brain capillaries using the histone deacetylase inhibitor (HDACi) valproic acid (VPA). METHODS Primary mouse brain capillary endothelial cells (BCECs) and brain capillaries isolated from mice injected intraperitoneally with VPA were examined using RT-qPCR and ELISA. Targeting to the BBB was performed by injecting monoclonal anti-TfR1 (Ri7217)-conjugated gold nanoparticles measured using ICP-MS. RESULTS In BCECs co-cultured with glial cells, Tfrc mRNA expression was significantly higher after 6 h VPA, returning to baseline after 24 h. In vivo Glut1 mRNA expression was significantly higher in males, but not females, receiving VPA, whereas Cd98hc mRNA expression was unaffected by VPA. TfR1 increased significantly in vivo after VPA, whereas GLUT1 and CD98hc were unchanged. The uptake of anti-TfR1-conjugated nanoparticles was unaltered by VPA despite upregulated TfR expression. CONCLUSIONS VPA upregulates TfR1 in brain endothelium in vivo and in vitro. VPA does not increase GLUT1 and CD98hc proteins. The increase in TfR1 does not result in higher anti-TfR1 antibody targetability, suggesting targeting sufficiently occurs with available transferrin receptors without further contribution from accessory VPA-induced TfR1.
Collapse
Affiliation(s)
- Steinunn Sara Helgudóttir
- Neurobiology Research and Drug Delivery (NRD), Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (S.S.H.); (L.G.R.); (C.L.M.R.)
| | - Kasper Bendix Johnsen
- Section for Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, 2800 Lyngby, Denmark;
| | - Lisa Greve Routhe
- Neurobiology Research and Drug Delivery (NRD), Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (S.S.H.); (L.G.R.); (C.L.M.R.)
| | - Charlotte Laurfelt Munch Rasmussen
- Neurobiology Research and Drug Delivery (NRD), Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (S.S.H.); (L.G.R.); (C.L.M.R.)
| | - Maj Schneider Thomsen
- Neurobiology Research and Drug Delivery (NRD), Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (S.S.H.); (L.G.R.); (C.L.M.R.)
| | - Torben Moos
- Neurobiology Research and Drug Delivery (NRD), Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (S.S.H.); (L.G.R.); (C.L.M.R.)
| |
Collapse
|
14
|
Barrera-Ocampo A. Monoclonal antibodies and aptamers: The future therapeutics for Alzheimer's disease. Acta Pharm Sin B 2024; 14:2795-2814. [PMID: 39027235 PMCID: PMC11252463 DOI: 10.1016/j.apsb.2024.03.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 07/20/2024] Open
Abstract
Alzheimer's disease (AD) is considered the most common and prevalent form of dementia of adult-onset with characteristic progressive impairment in cognition and memory. The cure for AD has not been found yet and the treatments available until recently were only symptomatic. Regardless of multidisciplinary approaches and efforts made by pharmaceutical companies, it was only in the past two years that new drugs were approved for the treatment of the disease. Amyloid beta (Aβ) immunotherapy is at the core of this therapy, which is one of the most innovative approaches looking to change the course of AD. This technology is based on synthetic peptides or monoclonal antibodies (mAb) to reduce Aβ levels in the brain and slow down the advance of neurodegeneration. Hence, this article reviews the state of the art about AD neuropathogenesis, the traditional pharmacologic treatment, as well as the modern active and passive immunization describing approved drugs, and drug prototypes currently under investigation in different clinical trials. In addition, future perspectives on immunotherapeutic strategies for AD and the rise of the aptamer technology as a non-immunogenic alternative to curb the disease progression are discussed.
Collapse
Affiliation(s)
- Alvaro Barrera-Ocampo
- Facultad de Ingeniería, Diseño y Ciencias Aplicadas, Departamento de Ciencias Farmacéuticas y Químicas, Grupo Natura, Universidad Icesi, Cali 760031, Colombia
| |
Collapse
|
15
|
Pinheiro FI, Araújo-Filho I, do Rego ACM, de Azevedo EP, Cobucci RN, Guzen FP. Hepatopancreatic metabolic disorders and their implications in the development of Alzheimer's disease and vascular dementia. Ageing Res Rev 2024; 96:102250. [PMID: 38417711 DOI: 10.1016/j.arr.2024.102250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/07/2024] [Accepted: 02/22/2024] [Indexed: 03/01/2024]
Abstract
Dementia has been faced with significant public health challenges and economic burdens that urges the need to develop safe and effective interventions. In recent years, an increasing number of studies have focused on the relationship between dementia and liver and pancreatic metabolic disorders that result in diseases such as diabetes, obesity, hypertension and dyslipidemia. Previous reports have shown that there is a plausible correlation between pathologies caused by hepatopancreatic dysfunctions and dementia. Glucose, insulin and IGF-1 metabolized in the liver and pancreas probably have an important influence on the pathophysiology of the most common dementias: Alzheimer's and vascular dementia. This current review highlights recent studies aimed at identifying convergent mechanisms, such as insulin resistance and other diseases, linked to altered hepatic and pancreatic metabolism, which are capable of causing brain changes that ultimately lead to dementia.
Collapse
Affiliation(s)
- Francisco I Pinheiro
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Department of Surgical, Federal University of Rio Grande do Norte, Natal 59010-180, Brazil; Institute of Education, Research and Innovation of the Liga Norte Rio-Grandense Against Cancer
| | - Irami Araújo-Filho
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Department of Surgical, Federal University of Rio Grande do Norte, Natal 59010-180, Brazil; Postgraduate Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, RN, Brazil
| | - Amália C M do Rego
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Institute of Education, Research and Innovation of the Liga Norte Rio-Grandense Against Cancer
| | - Eduardo P de Azevedo
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil
| | - Ricardo N Cobucci
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Postgraduate Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, RN, Brazil; Postgraduate Program in Science Applied to Women`s Health, Medical School, Federal University of Rio Grande do Norte (UFRN), Natal, RN, Brazil
| | - Fausto P Guzen
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Postgraduate Program in Health and Society, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró, Brazil; Postgraduate Program in Physiological Sciences, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró, Brazil.
| |
Collapse
|
16
|
Abdullahi A, Wong TW, Ng SS. Understanding the mechanisms of disease modifying effects of aerobic exercise in people with Alzheimer's disease. Ageing Res Rev 2024; 94:102202. [PMID: 38272266 DOI: 10.1016/j.arr.2024.102202] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/06/2023] [Accepted: 01/17/2024] [Indexed: 01/27/2024]
Abstract
Alzheimer's disease (AD) is a very disabling disease. Pathologically, it is characterized by the presence of amyloid plaques and neurofibrillary tangles in the brain that results in neurodegeneration. Its clinical manifestations include progressive memory impairment, language decline and difficulty in carrying out activities of daily living (ADL). The disease is managed using interventions such as pharmacological interventions and aerobic exercise. Use of aerobic exercise has shown some promises in reducing the risk of developing AD, and improving cognitive function and the ability to carry out both basic and instrumental ADL. Although, the mechanisms through which aerobic exercise improves AD are poorly understood, improvement in vascular function, brain glucose metabolism and cardiorespiratory fitness, increase in antioxidant capacity and haemoglobin level, amelioration of immune-related and inflammatory responses, modulation of concentration of circulating Neurotrophins and peptides and decrease in concentration of tau protein and cortisol level among others seem to be the possible mechanisms. Therefore, understanding these mechanisms is important to help characterize the dose and the nature of the aerobic exercise to be given. In addition, they may also help in finding ways to optimize other interventions such as the pharmacological interventions. However, more quality studies are needed to verify the mechanisms.
Collapse
Affiliation(s)
- Auwal Abdullahi
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, China
| | - Thomson Wl Wong
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, China
| | - Shamay Sm Ng
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, China.
| |
Collapse
|
17
|
Caminiti SP, De Francesco S, Tondo G, Galli A, Redolfi A, Perani D, the Alzheimer's Disease Neuroimaging Initiative. FDG-PET markers of heterogeneity and different risk of progression in amnestic MCI. Alzheimers Dement 2024; 20:159-172. [PMID: 37505996 PMCID: PMC10962797 DOI: 10.1002/alz.13385] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/19/2023] [Accepted: 06/12/2023] [Indexed: 07/30/2023]
Abstract
INTRODUCTION Amnestic mild cognitive impairment (aMCI) is emerging as a heterogeneous condition. METHODS We looked at a cohort of N = 207 aMCI subjects, with baseline fluorodeoxyglucose positron emission tomography (FDG-PET), T1 magnetic resonance imaging, cerebrospinal fluid (CSF), apolipoprotein E (APOE), and neuropsychological assessment. An algorithm based on FDG-PET hypometabolism classified each subject into subtypes, then compared biomarker measures and clinical progression. RESULTS Three subtypes emerged: hippocampal sparing-cortical hypometabolism, associated with younger age and the highest level of Alzheimer's disease (AD)-CSF pathology; hippocampal/cortical hypometabolism, associated with a high percentage of APOE ε3/ε4 or ε4/ε4 carriers; medial-temporal hypometabolism, characterized by older age, the lowest AD-CSF pathology, the most severe hippocampal atrophy, and a benign course. Within the whole cohort, the severity of temporo-parietal hypometabolism, correlated with AD-CSF pathology and marked the rate of progression of cognitive decline. DISCUSSION FDG-PET can distinguish clinically comparable aMCI at single-subject level with different risk of progression to AD dementia or stability. The obtained results can be useful for the optimization of pharmacological trials and automated-classification models. HIGHLIGHTS Algorithm based on FDG-PET hypometabolism demonstrates distinct subtypes across aMCI; Three different subtypes show heterogeneous biological profiles and risk of progression; The cortical hypometabolism is associated with AD pathology and cognitive decline; MTL hypometabolism is associated with the lowest conversion rate and CSF-AD pathology.
Collapse
Affiliation(s)
- Silvia Paola Caminiti
- Vita‐Salute San Raffaele UniversityMilanItaly
- Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Silvia De Francesco
- Laboratory of NeuroinformaticsIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Giacomo Tondo
- Vita‐Salute San Raffaele UniversityMilanItaly
- Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Alice Galli
- Vita‐Salute San Raffaele UniversityMilanItaly
- Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Alberto Redolfi
- Laboratory of NeuroinformaticsIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Daniela Perani
- Vita‐Salute San Raffaele UniversityMilanItaly
- Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
| | | |
Collapse
|
18
|
Milos T, Rojo D, Nedic Erjavec G, Konjevod M, Tudor L, Vuic B, Svob Strac D, Uzun S, Mimica N, Kozumplik O, Barbas C, Zarkovic N, Pivac N, Nikolac Perkovic M. Metabolic profiling of Alzheimer's disease: Untargeted metabolomics analysis of plasma samples. Prog Neuropsychopharmacol Biol Psychiatry 2023; 127:110830. [PMID: 37454721 DOI: 10.1016/j.pnpbp.2023.110830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 06/07/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Alzheimer's disease (AD) is often not recognized or is diagnosed very late, which significantly reduces the effectiveness of available pharmacological treatments. Metabolomic analyzes have great potential for improving existing knowledge about the pathogenesis and etiology of AD and represent a novel approach towards discovering biomarkers that could be used for diagnosis, prognosis, and therapy monitoring. In this study, we applied the untargeted metabolomic approach to investigate the changes in biochemical pathways related to AD pathology. We used gas chromatography and liquid chromatography coupled to mass spectrometry (GC-MS and LC-MS, respectively) to identify metabolites whose levels have changed in subjects with AD diagnosis (N = 40) compared to healthy controls (N = 40) and individuals with mild cognitive impairment (MCI, N = 40). The GC-MS identified significant differences between groups in levels of metabolites belonging to the classes of benzene and substituted derivatives, carboxylic acids and derivatives, fatty acyls, hydroxy acids and derivatives, keto acids and derivatives, and organooxygen compounds. Most of the compounds identified by the LC-MS were various fatty acyls, glycerolipids and glycerophospholipids. All of these compounds were decreased in AD patients and in subjects with MCI compared to healthy controls. The results of the study indicate disturbed metabolism of lipids and amino acids and an imbalance of metabolites involved in energy metabolism in individuals diagnosed with AD, compared to healthy controls and MCI subjects.
Collapse
Affiliation(s)
- Tina Milos
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia.
| | - David Rojo
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities Madrid, Spain.
| | | | - Marcela Konjevod
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia.
| | - Lucija Tudor
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia.
| | - Barbara Vuic
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia.
| | | | - Suzana Uzun
- School of Medicine, University of Zagreb, Zagreb, Croatia; Department for Biological Psychiatry and Psychogeriatrics, University Psychiatric Hospital Vrapče, Zagreb, Croatia.
| | - Ninoslav Mimica
- Department for Biological Psychiatry and Psychogeriatrics, University Psychiatric Hospital Vrapče, Zagreb, Croatia.
| | - Oliver Kozumplik
- Department for Biological Psychiatry and Psychogeriatrics, University Psychiatric Hospital Vrapče, Zagreb, Croatia.
| | - Coral Barbas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities Madrid, Spain.
| | - Neven Zarkovic
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia.
| | - Nela Pivac
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia; University of Applied Sciences Hrvatsko Zagorje Krapina, Krapina, Croatia.
| | | |
Collapse
|
19
|
Honea RA, Hunt S, Lepping RJ, Vidoni ED, Morris JK, Watts A, Michaelis E, Burns JM, Swerdlow RH. Alzheimer's disease cortical morphological phenotypes are associated with TOMM40'523-APOE haplotypes. Neurobiol Aging 2023; 132:131-144. [PMID: 37804609 PMCID: PMC10763175 DOI: 10.1016/j.neurobiolaging.2023.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 10/09/2023]
Abstract
Both the APOE ε4 and TOMM40 rs10524523 ("523") genes have been associated with risk for Alzheimer's disease (AD) and neuroimaging biomarkers of AD. No studies have investigated the relationship of TOMM40'523-APOE ε4 on the structural complexity of the brain in AD individuals. We quantified brain morphology and multiple cortical attributes in individuals with mild cognitive impairment (MCI) and AD, then tested whether APOE ε4 or TOMM40 poly-T genotypes were related to AD morphological biomarkers in cognitively unimpaired (CU) and MCI/AD individuals. We identified several AD-specific phenotypes in brain morphology and found that TOMM40 poly-T short alleles are associated with early, AD-specific brain morphological differences in healthy aging. We observed decreased cortical thickness, sulcal depth, and fractal dimension in CU individuals with the poly-T short alleles. Moreover, in MCI/AD participants, the APOE ε4 (TOMM40 L) individuals had a higher rate of gene-related morphological markers indicative of AD. Our data suggest that TOMM40'523 is associated with early brain structure variations in the precuneus, temporal, and limbic cortices.
Collapse
Affiliation(s)
- Robyn A Honea
- University of Kansas Alzheimer's Disease Center, University of Kansas School of Medicine, Kansas City, KS, USA; Department of Neurology, University of Kansas School of Medicine, Kansas City, KS, USA.
| | - Suzanne Hunt
- University of Kansas Alzheimer's Disease Center, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Rebecca J Lepping
- University of Kansas Alzheimer's Disease Center, University of Kansas School of Medicine, Kansas City, KS, USA; Hoglund Biomedical Imaging Center, University of Kansas Medical Center, Kansas City, KS, USA; Department of Neurology, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Eric D Vidoni
- University of Kansas Alzheimer's Disease Center, University of Kansas School of Medicine, Kansas City, KS, USA; Department of Neurology, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Jill K Morris
- University of Kansas Alzheimer's Disease Center, University of Kansas School of Medicine, Kansas City, KS, USA; Department of Neurology, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Amber Watts
- University of Kansas Alzheimer's Disease Center, University of Kansas School of Medicine, Kansas City, KS, USA; Department of Psychology, University of Kansas, Lawrence, KS, USA
| | - Elias Michaelis
- University of Kansas Alzheimer's Disease Center, University of Kansas School of Medicine, Kansas City, KS, USA; Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, USA
| | - Jeffrey M Burns
- University of Kansas Alzheimer's Disease Center, University of Kansas School of Medicine, Kansas City, KS, USA; Department of Neurology, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Russell H Swerdlow
- University of Kansas Alzheimer's Disease Center, University of Kansas School of Medicine, Kansas City, KS, USA; Department of Neurology, University of Kansas School of Medicine, Kansas City, KS, USA
| |
Collapse
|
20
|
Ito S, Yagi R, Ogata S, Masuda T, Saito T, Saido T, Ohtsuki S. Proteomic alterations in the brain and blood-brain barrier during brain Aβ accumulation in an APP knock-in mouse model of Alzheimer's disease. Fluids Barriers CNS 2023; 20:66. [PMID: 37705104 PMCID: PMC10500766 DOI: 10.1186/s12987-023-00466-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 09/02/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Blood-brain barrier (BBB) dysfunction is supposed to be an early event in the development of Alzheimer's disease (AD). This study aimed to investigate the relationship between BBB alterations and AD progression in terms of amyloid-β peptide (Aβ) accumulation in the brains of humanized amyloid precursor protein knock-in (APP-KI) mice. METHODS Brain Aβ accumulation was examined using immunohistochemical analysis. Alterations in differentially expressed proteins were determined using sequential window acquisition of all theoretical fragment ion mass spectroscopy (SWATH-MS)-based quantitative proteomics, and Metascape, STRING, Gene Ontology, and KEGG were used for network analyses of altered biological pathways and processes. Statistical significance was determined using the unpaired two-tailed Student's t-test and Welch's t-test for two groups and one-way analysis of variance followed by Tukey's test for more than two groups. Correlations between two groups were determined using Pearson's correlation analysis. RESULTS Brain Aβ accumulation in APP-KI mice was detectable at 2 months, increased significantly at 5 months, and remained elevated at 12 months of age. The levels of differentially expressed proteins in isolated brain capillaries were higher in younger mice, whereas those in the brain were higher in older mice. Network analyses indicated changes in basement membrane-associated and ribosomal proteins in the brain capillaries. There were no significant changes in key proteins involved in drug or Aβ transport at the BBB. In contrast, solute carrier transporter levels in astrocytes, microglia, and neurons were altered in the brain of older mice. Moreover, the levels of the lipid transporters Apoe and Apoj were upregulated in both the brain and isolated brain capillaries after Aβ accumulation. CONCLUSIONS Our results suggest that changes in the brain occurred after advanced Aβ accumulation, whereas initial Aβ accumulation was sufficient to cause alterations in the BBB. These findings may help elucidate the role of BBB alterations in AD progression and predict the distribution of drugs across the BBB in the brain of patients with AD.
Collapse
Affiliation(s)
- Shingo Ito
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan.
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan.
| | - Ryotaro Yagi
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Seiryo Ogata
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Takeshi Masuda
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Takaomi Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| |
Collapse
|
21
|
Hashemi R, Vahabi Z, Rasekhi H, Shiraseb F, Amini M. Dietary patterns and the risk of Alzheimer's disease in an elderly Iranian population: a case-control study. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2023; 42:56. [PMID: 37322540 DOI: 10.1186/s41043-023-00398-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/09/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND An increasing number of studies in Western countries have shown that healthy eating patterns have a protective effect against cognitive decline and dementia, however, information about this relationship among non-western populations with different cultural environments is scarce. The present study investigated the association between dietary patterns (DPs) and cognitive function in the Iranian elderly. METHODS In this case-control study, the data of 290 elderly people in two groups of case and control (Mean age in case: 74.2 ± 8.6, in control: 67.3 ± 7.3 year) were analyzed. Two DPs of healthy and unhealthy were extracted from a 142-item dish-based food frequency questionnaire, and patterns driven by principal components analysis (PCA) of 25 food groups. Multivariate binary logistic regression calculated the odds ratio (OR) of cognitive impairment with adjustment for potential confounding factors. RESULTS A healthy DP, characterized by high consumption of fruits and vegetables, legumes, and nuts, was related to a decrease in the odds of Alzheimer's disease in Iranian elderly people. Also, moderate adherence to an unhealthy food pattern was associated with an increase in the probability of the disease; however, the association was not statistically significant. CONCLUSION In this elderly population, a healthy eating pattern was associated with reducing the risk of Alzheimer's disease. Further prospective studies are recommended.
Collapse
Affiliation(s)
- Rezvan Hashemi
- Department of Geriatric Medicine, Ziaeian Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Vahabi
- Geriatric Department, Tehran University of Medical Sciences, Tehran, Iran
- Neurology Division, CNS Department, Western University, London, ON, Canada
| | - Hamid Rasekhi
- Department of Nutrition Research, National Nutrition and Food Technology Research Institute and Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farideh Shiraseb
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Amini
- Department of Nutrition Research, National Nutrition and Food Technology Research Institute and Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Jett S, Boneu C, Zarate C, Carlton C, Kodancha V, Nerattini M, Battista M, Pahlajani S, Williams S, Dyke JP, Mosconi L. Systematic review of 31P-magnetic resonance spectroscopy studies of brain high energy phosphates and membrane phospholipids in aging and Alzheimer's disease. Front Aging Neurosci 2023; 15:1183228. [PMID: 37273652 PMCID: PMC10232902 DOI: 10.3389/fnagi.2023.1183228] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/02/2023] [Indexed: 06/06/2023] Open
Abstract
Many lines of evidence suggest that mitochondria have a central role in aging-related neurodegenerative diseases, such as Alzheimer's disease (AD). Mitochondrial dysfunction, cerebral energy dysmetabolism and oxidative damage increase with age, and are early event in AD pathophysiology and may precede amyloid beta (Aβ) plaques. In vivo probes of mitochondrial function and energy metabolism are therefore crucial to characterize the bioenergetic abnormalities underlying AD risk, and their relationship to pathophysiology and cognition. A majority of the research conducted in humans have used 18F-fluoro-deoxygluose (FDG) PET to image cerebral glucose metabolism (CMRglc), but key information regarding oxidative phosphorylation (OXPHOS), the process which generates 90% of the energy for the brain, cannot be assessed with this method. Thus, there is a crucial need for imaging tools to measure mitochondrial processes and OXPHOS in vivo in the human brain. 31Phosphorus-magnetic resonance spectroscopy (31P-MRS) is a non-invasive method which allows for the measurement of OXPHOS-related high-energy phosphates (HEP), including phosphocreatine (PCr), adenosine triphosphate (ATP), and inorganic phosphate (Pi), in addition to potential of hydrogen (pH), as well as components of phospholipid metabolism, such as phosphomonoesters (PMEs) and phosphodiesters (PDEs). Herein, we provide a systematic review of the existing literature utilizing the 31P-MRS methodology during the normal aging process and in patients with mild cognitive impairment (MCI) and AD, with an additional focus on individuals at risk for AD. We discuss the strengths and limitations of the technique, in addition to considering future directions toward validating the use of 31P-MRS measures as biomarkers for the early detection of AD.
Collapse
Affiliation(s)
- Steven Jett
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Camila Boneu
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Camila Zarate
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Caroline Carlton
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Vibha Kodancha
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Matilde Nerattini
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Michael Battista
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Silky Pahlajani
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
- Department of Radiology, Weill Cornell Medical College, New York, NY, United States
| | - Schantel Williams
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Jonathan P. Dyke
- Department of Radiology, Weill Cornell Medical College, New York, NY, United States
| | - Lisa Mosconi
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
- Department of Radiology, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
23
|
Afsar A, Chacon Castro MDC, Soladogun AS, Zhang L. Recent Development in the Understanding of Molecular and Cellular Mechanisms Underlying the Etiopathogenesis of Alzheimer's Disease. Int J Mol Sci 2023; 24:7258. [PMID: 37108421 PMCID: PMC10138573 DOI: 10.3390/ijms24087258] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that leads to dementia and patient death. AD is characterized by intracellular neurofibrillary tangles, extracellular amyloid beta (Aβ) plaque deposition, and neurodegeneration. Diverse alterations have been associated with AD progression, including genetic mutations, neuroinflammation, blood-brain barrier (BBB) impairment, mitochondrial dysfunction, oxidative stress, and metal ion imbalance.Additionally, recent studies have shown an association between altered heme metabolism and AD. Unfortunately, decades of research and drug development have not produced any effective treatments for AD. Therefore, understanding the cellular and molecular mechanisms underlying AD pathology and identifying potential therapeutic targets are crucial for AD drug development. This review discusses the most common alterations associated with AD and promising therapeutic targets for AD drug discovery. Furthermore, it highlights the role of heme in AD development and summarizes mathematical models of AD, including a stochastic mathematical model of AD and mathematical models of the effect of Aβ on AD. We also summarize the potential treatment strategies that these models can offer in clinical trials.
Collapse
Affiliation(s)
| | | | | | - Li Zhang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
24
|
Cacciaglia R, Operto G, Falcón C, de Echavarri-Gómez JMG, Sánchez-Benavides G, Brugulat-Serrat A, Milà-Alomà M, Blennow K, Zetterberg H, Molinuevo JL, Suárez-Calvet M, Gispert JD. Genotypic effects of APOE-ε4 on resting-state connectivity in cognitively intact individuals support functional brain compensation. Cereb Cortex 2023; 33:2748-2760. [PMID: 35753703 PMCID: PMC10016049 DOI: 10.1093/cercor/bhac239] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 11/12/2022] Open
Abstract
The investigation of resting-state functional connectivity (rsFC) in asymptomatic individuals at genetic risk for Alzheimer's disease (AD) enables discovering the earliest brain alterations in preclinical stages of the disease. The APOE-ε4 variant is the major genetic risk factor for AD, and previous studies have reported rsFC abnormalities in carriers of the ε4 allele. Yet, no study has assessed APOE-ε4 gene-dose effects on rsFC measures, and only a few studies included measures of cognitive performance to aid a clinical interpretation. We assessed the impact of APOE-ε4 on rsFC in a sample of 429 cognitively unimpaired individuals hosting a high number of ε4 homozygotes (n = 58), which enabled testing different models of genetic penetrance. We used independent component analysis and found a reduced rsFC as a function of the APOE-ε4 allelic load in the temporal default-mode and the medial temporal networks, while recessive effects were found in the extrastriate and limbic networks. Some of these results were replicated in a subsample with negative amyloid markers. Interaction with cognitive data suggests that such a network reorganization may support cognitive performance in the ε4-homozygotes. Our data indicate that APOE-ε4 shapes the functional architecture of the resting brain and favor the idea of a network-based functional compensation.
Collapse
Affiliation(s)
- Raffaele Cacciaglia
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), 08005 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 28089 Madrid, Spain
| | - Grégory Operto
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), 08005 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 28089 Madrid, Spain
| | - Carles Falcón
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), 08005 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBERBBN), 28089 Madrid, Spain
| | - José Maria González de Echavarri-Gómez
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), 08005 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 28089 Madrid, Spain
| | - Gonzalo Sánchez-Benavides
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), 08005 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 28089 Madrid, Spain
| | - Anna Brugulat-Serrat
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), 08005 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 28089 Madrid, Spain
| | - Marta Milà-Alomà
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), 08005 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 28089 Madrid, Spain
- Universitat Pompeu Fabra, 08002 Barcelona, Spain
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, 41390 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 41390 Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, 41390 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 41390 Mölndal, Sweden
- UK Dementia Research Institute at UCL, WC1E 6BT London, United Kingdom
- Department of Neurodegenerative Disease, UCL Institute of Neurology, WC1N 3BG London, United Kingdom
- Honk Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), 08005 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 28089 Madrid, Spain
- Servei de Neurologia, Hospital del Mar, Barcelona, Spain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), 08005 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBERBBN), 28089 Madrid, Spain
- Universitat Pompeu Fabra, 08002 Barcelona, Spain
| |
Collapse
|
25
|
Jett S, Dyke JP, Boneu Yepez C, Zarate C, Carlton C, Schelbaum E, Jang G, Pahlajani S, Williams S, Diaz Brinton R, Mosconi L. Effects of sex and APOE ε4 genotype on brain mitochondrial high-energy phosphates in midlife individuals at risk for Alzheimer's disease: A 31Phosphorus MR spectroscopy study. PLoS One 2023; 18:e0281302. [PMID: 36787293 PMCID: PMC9928085 DOI: 10.1371/journal.pone.0281302] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 01/19/2023] [Indexed: 02/15/2023] Open
Abstract
Age, female sex, and APOE epsilon 4 (APOE4) genotype are the three greatest risk factors for late-onset Alzheimer's disease (AD). The convergence of these risks creates a hypometabolic AD-risk profile unique to women, which may help explain their higher lifetime risk of AD. Less is known about APOE4 effects in men, although APOE4 positive men also experience an increased AD risk. This study uses 31Phosphorus Magnetic Resonance Spectroscopy (31P-MRS) to examine effects of sex and APOE4 status on brain high-energy phosphates [adenosine triphosphate (ATP), phosphocreatine (PCr), inorganic phosphate (Pi)] and membrane phospholipids [phosphomonoesters (PME), phosphodiesters (PDE)] in 209 cognitively normal individuals at risk for AD, ages 40-65, 80% female, 46% APOE4 carriers (APOE4+). Women exhibited lower PCr/ATP and PCr/Pi levels than men in AD-vulnerable regions, including frontal, posterior cingulate, lateral and medial temporal cortex (multi-variable adjusted p≤0.037). The APOE4+ group exhibited lower PCr/ATP and PCr/Pi in frontal regions as compared to non-carriers (APOE4-) (multi-variable adjusted p≤0.005). Sex by APOE4 status interactions were observed in frontal regions (multi-variable adjusted p≤0.046), where both female groups and APOE4+ men exhibited lower PCr/ATP and PCr/Pi than APOE4- men. Among men, APOE4 homozygotes exhibited lower frontal PCr/ATP than heterozygotes and non-carriers. There were no significant effects of sex or APOE4 status on Pi/ATP and PME/PDE measures. Among midlife individuals at risk for AD, women exhibit lower PCr/ATP (e.g. higher ATP utilization) and lower PCr/Pi (e.g. higher energy demand) than age-controlled men, independent of APOE4 status. However, a double dose of APOE4 allele shifted men's brains to a similar metabolic range as women's brains. Examination of brain metabolic heterogeneity can support identification of AD-specific pathways within at-risk subgroups, further advancing both preventive and precision medicine for AD.
Collapse
Affiliation(s)
- Steven Jett
- Department of Neurology, Weill Cornell Medicine, New York, New York, United States of America
| | - Jonathan P. Dyke
- Department of Radiology, Weill Cornell Medicine, New York, New York, United States of America
| | - Camila Boneu Yepez
- Department of Neurology, Weill Cornell Medicine, New York, New York, United States of America
| | - Camila Zarate
- Department of Neurology, Weill Cornell Medicine, New York, New York, United States of America
| | - Caroline Carlton
- Department of Neurology, Weill Cornell Medicine, New York, New York, United States of America
| | - Eva Schelbaum
- Department of Neurology, Weill Cornell Medicine, New York, New York, United States of America
| | - Grace Jang
- Department of Neurology, Weill Cornell Medicine, New York, New York, United States of America
| | - Silky Pahlajani
- Department of Neurology, Weill Cornell Medicine, New York, New York, United States of America
- Department of Radiology, Weill Cornell Medicine, New York, New York, United States of America
| | - Schantel Williams
- Department of Neurology, Weill Cornell Medicine, New York, New York, United States of America
| | - Roberta Diaz Brinton
- Department of Pharmacology, University of Arizona, Tucson, Arizona, United States of America
- Department of Neurology, University of Arizona, Tucson, Arizona, United States of America
| | - Lisa Mosconi
- Department of Neurology, Weill Cornell Medicine, New York, New York, United States of America
- Department of Radiology, Weill Cornell Medicine, New York, New York, United States of America
| |
Collapse
|
26
|
Sousa JA, Bernardes C, Bernardo-Castro S, Lino M, Albino I, Ferreira L, Brás J, Guerreiro R, Tábuas-Pereira M, Baldeiras I, Santana I, Sargento-Freitas J. Reconsidering the role of blood-brain barrier in Alzheimer's disease: From delivery to target. Front Aging Neurosci 2023; 15:1102809. [PMID: 36875694 PMCID: PMC9978015 DOI: 10.3389/fnagi.2023.1102809] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
The existence of a selective blood-brain barrier (BBB) and neurovascular coupling are two unique central nervous system vasculature features that result in an intimate relationship between neurons, glia, and blood vessels. This leads to a significant pathophysiological overlap between neurodegenerative and cerebrovascular diseases. Alzheimer's disease (AD) is the most prevalent neurodegenerative disease whose pathogenesis is still to be unveiled but has mostly been explored under the light of the amyloid-cascade hypothesis. Either as a trigger, bystander, or consequence of neurodegeneration, vascular dysfunction is an early component of the pathological conundrum of AD. The anatomical and functional substrate of this neurovascular degeneration is the BBB, a dynamic and semi-permeable interface between blood and the central nervous system that has consistently been shown to be defective. Several molecular and genetic changes have been demonstrated to mediate vascular dysfunction and BBB disruption in AD. The isoform ε4 of Apolipoprotein E is at the same time the strongest genetic risk factor for AD and a known promoter of BBB dysfunction. Low-density lipoprotein receptor-related protein 1 (LRP-1), P-glycoprotein, and receptor for advanced glycation end products (RAGE) are examples of BBB transporters implicated in its pathogenesis due to their role in the trafficking of amyloid-β. This disease is currently devoid of strategies that change the natural course of this burdening illness. This unsuccess may partly be explained by our misunderstanding of the disease pathogenesis and our inability to develop drugs that are effectively delivered to the brain. BBB may represent a therapeutic opportunity as a target itself or as a therapeutic vehicle. In this review, we aim to explore the role of BBB in the pathogenesis of AD including the genetic background and detail how it can be targeted in future therapeutic research.
Collapse
Affiliation(s)
- João André Sousa
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Catarina Bernardes
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Sara Bernardo-Castro
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Miguel Lino
- Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Inês Albino
- Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Lino Ferreira
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - José Brás
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Rita Guerreiro
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Miguel Tábuas-Pereira
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Inês Baldeiras
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - Isabel Santana
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - João Sargento-Freitas
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
27
|
Solas M, Zamarbide M, Ardanaz CG, Ramírez MJ, Pérez-Mediavilla A. The Cognitive Improvement and Alleviation of Brain Hypermetabolism Caused by FFAR3 Ablation in Tg2576 Mice Is Persistent under Diet-Induced Obesity. Int J Mol Sci 2022; 23:13591. [PMID: 36362376 PMCID: PMC9654726 DOI: 10.3390/ijms232113591] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/28/2022] [Accepted: 11/03/2022] [Indexed: 11/10/2022] Open
Abstract
Obesity and aging are becoming increasingly prevalent across the globe. It has been established that aging is the major risk factor for Alzheimer's disease (AD), and it is becoming increasingly evident that obesity and the associated insulin resistance are also notably relevant risk factors. The biological plausibility of the link between high adiposity, insulin resistance, and dementia is central for understanding AD etiology, and to form bases for prevention efforts to decrease the disease burden. Several studies have demonstrated a strong association between short chain fatty acid receptor FFAR3 and insulin sensitivity. Interestingly, it has been recently established that FFAR3 mRNA levels are increased in early stages of the AD pathology, indicating that FFAR3 could play a key role in AD onset and progression. Indeed, in the present study we demonstrate that the ablation of the Ffar3 gene in Tg2576 mice prevents the development of cognitive deficiencies in advanced stages of the disease. Notably, this cognitive improvement is also maintained upon a severe metabolic challenge such as the exposure to high-fat diet (HFD) feeding. Moreover, FFAR3 deletion restores the brain hypermetabolism displayed by Tg2576 mice. Collectively, these data postulate FFAR3 as a potential novel target for AD.
Collapse
Affiliation(s)
- Maite Solas
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Marta Zamarbide
- Neuroscience Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
| | - Carlos G. Ardanaz
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - María J. Ramírez
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Alberto Pérez-Mediavilla
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Neuroscience Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain
| |
Collapse
|
28
|
Kumar V, Kim SH, Bishayee K. Dysfunctional Glucose Metabolism in Alzheimer’s Disease Onset and Potential Pharmacological Interventions. Int J Mol Sci 2022; 23:ijms23179540. [PMID: 36076944 PMCID: PMC9455726 DOI: 10.3390/ijms23179540] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/05/2022] [Accepted: 08/21/2022] [Indexed: 12/04/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common age-related dementia. The alteration in metabolic characteristics determines the prognosis. Patients at risk show reduced glucose uptake in the brain. Additionally, type 2 diabetes mellitus increases the risk of AD with increasing age. Therefore, changes in glucose uptake in the cerebral cortex may predict the histopathological diagnosis of AD. The shifts in glucose uptake and metabolism, insulin resistance, oxidative stress, and abnormal autophagy advance the pathogenesis of AD syndrome. Here, we summarize the role of altered glucose metabolism in type 2 diabetes for AD prognosis. Additionally, we discuss diagnosis and potential pharmacological interventions for glucose metabolism defects in AD to encourage the development of novel therapeutic methods.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - So-Hyeon Kim
- Biomedical Science Core-Facility, Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan 31151, Korea
| | - Kausik Bishayee
- Biomedical Science Core-Facility, Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan 31151, Korea
- Correspondence: or
| |
Collapse
|
29
|
Lin Y, Dai X, Zhang J, Chen X. Metformin alleviates the depression-like behaviors of elderly apoE4 mice via improving glucose metabolism and mitochondrial biogenesis. Behav Brain Res 2022; 423:113772. [PMID: 35090900 DOI: 10.1016/j.bbr.2022.113772] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/28/2021] [Accepted: 01/22/2022] [Indexed: 12/25/2022]
Abstract
Apolipoprotein E4 (apoE4) is closely related to late-onset depression (LOD). In addition, the benefits of metformin treatment of depression have been documented in a range of rodent studies and human trials, but few studies have probed into the effect of metformin on and the related mechanism in depressed elderly mice, especially in those APOE4 carriers. Here, we treated 13-month-old apoE3-targeted replacement (TR) and apoE4-TR mice with an intragastric administration of metformin (300 mg/kg/d) or normal saline for 5 months. We found that metformin exerted antidepressant effects on apoE4 mice, including reduced immobility time in TST and FST, and increased ratios of time and distance in the central area of OFT. Importantly, compared with apoE3 mice, apoE4 mice showed a higher expression of lactate dehydrogenase (LDH) and pyruvate dehydrogenase kinase (PDK1 and PDK4) in the hippocampus. The increased LDH level was rescued by metformin treatment. Moreover, the metformin administration increased the levels of transcriptional factor NRF-1 and TFAM, mtDNA, and most mitochondrial complex subunits in apoE-TR mice. Furthermore, it upregulated the expressions of antioxidant enzymes, such as MnSOD, GPX1, and GSR1/2. Interestingly, apoE4 blunted the hypoglycemic effect of metformin in aged mice. These data suggest that metformin ameliorates the depression-like behaviors probably by improving glucose metabolism and mitochondria biogenesis in the hippocampus of aged apoE4 mice. These findings imply that chronic metformin treatment can improve apoE4-mediated LOD, providing mechanistic insights for apoE4- and age-based depression prevention and therapy.
Collapse
Affiliation(s)
- Yingbin Lin
- Department of Neurology and Geriatrics, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian 350001, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350001, China
| | - Xiaoman Dai
- Department of Neurology and Geriatrics, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian 350001, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350001, China; Fujian Key Laboratory of Vascular Aging, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350001, China
| | - Jing Zhang
- Department of Neurology and Geriatrics, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian 350001, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350001, China; Fujian Key Laboratory of Vascular Aging, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350001, China.
| | - Xiaochun Chen
- Department of Neurology and Geriatrics, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian 350001, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350001, China.
| |
Collapse
|
30
|
Brain Metabolic Alterations in Alzheimer's Disease. Int J Mol Sci 2022; 23:ijms23073785. [PMID: 35409145 PMCID: PMC8998942 DOI: 10.3390/ijms23073785] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
The brain is one of the most energy-consuming organs in the body. Satisfying such energy demand requires compartmentalized, cell-specific metabolic processes, known to be complementary and intimately coupled. Thus, the brain relies on thoroughly orchestrated energy-obtaining agents, processes and molecular features, such as the neurovascular unit, the astrocyte-neuron metabolic coupling, and the cellular distribution of energy substrate transporters. Importantly, early features of the aging process are determined by the progressive perturbation of certain processes responsible for adequate brain energy supply, resulting in brain hypometabolism. These age-related brain energy alterations are further worsened during the prodromal stages of neurodegenerative diseases, namely Alzheimer's disease (AD), preceding the onset of clinical symptoms, and are anatomically and functionally associated with the loss of cognitive abilities. Here, we focus on concrete neuroenergetic features such as the brain's fueling by glucose and lactate, the transporters and vascular system guaranteeing its supply, and the metabolic interactions between astrocytes and neurons, and on its neurodegenerative-related disruption. We sought to review the principles underlying the metabolic dimension of healthy and AD brains, and suggest that the integration of these concepts in the preventive, diagnostic and treatment strategies for AD is key to improving the precision of these interventions.
Collapse
|
31
|
Liu Y, Han PR, Hu H, Wang ZT, Guo Y, Ou YN, Cao XP, Tan L, Yu JT. A Multi-Dimensional Comparison of Alzheimer's Disease Neurodegenerative Biomarkers. J Alzheimers Dis 2022; 87:197-209. [PMID: 35275546 DOI: 10.3233/jad-215724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND In the 2018 AT(N) framework, neurodegenerative (N) biomarkers plays an essential role in the research and staging of Alzheimer's disease (AD); however, the different choice of N may result in discordances. OBJECTIVE We aimed to compare different potential N biomarkers. METHODS We examined these N biomarkers among 1,238 participants from Alzheimer's Disease Neuroimaging Initiative (ADNI) in their 1) diagnostic utility, 2) cross-sectional and longitudinal correlations between different N biomarkers and clinical variables, and 3) the conversion risk of different N profiles. RESULTS Six neurodegenerative biomarkers changed significantly from preclinical AD, through prodromal AD to AD dementia stage, thus they were chosen as the candidate N biomarkers: hippocampal volume (HV), 18F-fluorodeoxyglucose-positron emission tomography (FDG-PET), cerebrospinal fluid (CSF), total tau (T-tau), plasma neurofilament light chain (NFL), CSF NFL, and CSF neurogranin (Ng). Results indicated that FDG-PET not only had the greatest diagnostic utility in differentiating AD from controls (area under the curve: FDG-PET, 0.922), but also had the strongest association with cognitive scores. Furthermore, FDG-PET positive group showed the fastest memory decline (hazard ratio: FDG-PET, 3.45), which was also true even in the presence of amyloid-β pathology. Moreover, we observed great discordances between three valuable N biomarkers (FDG-PET, HV, and T-tau). CONCLUSION These results underline the importance of using FDG-PET as N in terms of cognitive decline and AD conversion, followed by HV, and could be a great complement to the AT(N) framework.
Collapse
Affiliation(s)
- Ying Liu
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Pei-Ran Han
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yu Guo
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xi-Peng Cao
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing, China.,Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | | |
Collapse
|
32
|
Regy M, Dugravot A, Sabia S, Fayosse A, Mangin JF, Chupin M, Fischer C, Bouteloup V, Dufouil C, Chêne G, Paquet C, Hanseeuw B, Singh-Manoux A, Dumurgier J. Association of APOE ε4 with cerebral gray matter volumes in non-demented older adults: the MEMENTO cohort study. Neuroimage 2022; 250:118966. [PMID: 35122970 DOI: 10.1016/j.neuroimage.2022.118966] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 01/24/2022] [Accepted: 02/01/2022] [Indexed: 10/19/2022] Open
Abstract
Data on 2,045 non-demented individuals with memory complaints were drawn from the Memento cohort study to examine the association between Apolipoprotein E ε4 allele (APOE4) and regional brain gray matter volumes. Linear regression was used to examine the association of APOE4 and measures of regional gray matter volumes in cross-sectional analysis and change therein using longitudinal analyses based on two brain MRI performed at baseline and at two-year follow-up. Overall, in analyses adjusted for age, sex, and intracranial volume, the presence of APOE4 was associated with lower total gray matter volume at baseline and with a higher atrophy rate over the follow-up. The hippocampus and entorhinal cortex were the two gray matter regions most associated with APOE4. Further adjustment for cardiovascular risk factors had little impact on these associations. There was an interaction between age, APOE4 status and total brain volume atrophy rate, with evidence of an earlier age at onset of atrophy in hippocampal volume in APOE4 carriers compared to non-carriers. Those results are in accordance with the role of medial temporal structures in the greater risk of dementia observed in people carrying the APOE4 allele.
Collapse
Affiliation(s)
- Melina Regy
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative diseases, Paris, France; Université catholique de Louvain, Louvain, Belgium.
| | - Aline Dugravot
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative diseases, Paris, France
| | - Séverine Sabia
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative diseases, Paris, France; University College London, Department of Epidemiology and Public Health, London, United Kingdom
| | - Aurore Fayosse
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative diseases, Paris, France
| | - Jean-Francois Mangin
- Université Paris-Saclay, CEA, CNRS, CATI, NeuroSpin, Baobab, Gif sur Yvette, France
| | - Marie Chupin
- Université Paris-Saclay, CEA, CNRS, CATI, NeuroSpin, Baobab, Gif sur Yvette, France
| | - Clara Fischer
- Université Paris-Saclay, CEA, CNRS, CATI, NeuroSpin, Baobab, Gif sur Yvette, France
| | - Vincent Bouteloup
- Université de Bordeaux, Bordeaux, France; Pôle de Santé publique Centre Hospitalier Universitaire de Bordeaux, Inserm, UMR 1219, Inserm, CIC1401-EC, Bordeaux, France
| | - Carole Dufouil
- Université de Bordeaux, Bordeaux, France; Pôle de Santé publique Centre Hospitalier Universitaire de Bordeaux, Inserm, UMR 1219, Inserm, CIC1401-EC, Bordeaux, France
| | - Geneviève Chêne
- Université de Bordeaux, Bordeaux, France; Pôle de Santé publique Centre Hospitalier Universitaire de Bordeaux, Inserm, UMR 1219, Inserm, CIC1401-EC, Bordeaux, France
| | - Claire Paquet
- GHU APHP Nord Université de Paris Lariboisiere - Fernand Widal Paris, France; Université de Paris, INSERMU1144, Paris France
| | - Bernard Hanseeuw
- Université catholique de Louvain, Louvain, Belgium; Cliniques Universitaires Saint-Luc, Bruxelles, Belgium
| | - Archana Singh-Manoux
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative diseases, Paris, France; University College London, Department of Epidemiology and Public Health, London, United Kingdom
| | - Julien Dumurgier
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative diseases, Paris, France; GHU APHP Nord Université de Paris Lariboisiere - Fernand Widal Paris, France
| | | |
Collapse
|
33
|
Wang G, Zhou W, Kong D, Qu Z, Ba M, Hao J, Yao T, Dong Q, Su Y, Reiman EM, Caselli RJ, Chen K, Wang Y. Studying APOE ɛ4 Allele Dose Effects with a Univariate Morphometry Biomarker. J Alzheimers Dis 2022; 85:1233-1250. [PMID: 34924383 PMCID: PMC10498787 DOI: 10.3233/jad-215149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND A univariate neurodegeneration biomarker (UNB) based on MRI with strong statistical discrimination power would be highly desirable for studying hippocampal surface morphological changes associated with APOE ɛ4 genetic risk for AD in the cognitively unimpaired (CU) population. However, existing UNB work either fails to model large group variances or does not capture AD induced changes. OBJECTIVE We proposed a subspace decomposition method capable of exploiting a UNB to represent the hippocampal morphological changes related to the APOE ɛ4 dose effects among the longitudinal APOE ɛ4 homozygotes (HM, N = 30), heterozygotes (HT, N = 49) and non-carriers (NC, N = 61). METHODS Rank minimization mechanism combined with sparse constraint considering the local continuity of the hippocampal atrophy regions is used to extract group common structures. Based on the group common structures of amyloid-β (Aβ) positive AD patients and Aβ negative CU subjects, we identified the regions-of-interest (ROI), which reflect significant morphometry changes caused by the AD development. Then univariate morphometry index (UMI) is constructed from these ROIs. RESULTS The proposed UMI demonstrates a more substantial statistical discrimination power to distinguish the longitudinal groups with different APOE ɛ4 genotypes than the hippocampal volume measurements. And different APOE ɛ4 allele load affects the shrinkage rate of the hippocampus, i.e., HM genotype will cause the largest atrophy rate, followed by HT, and the smallest is NC. CONCLUSION The UMIs may capture the APOE ɛ4 risk allele-induced brain morphometry abnormalities and reveal the dose effects of APOE ɛ4 on the hippocampal morphology in cognitively normal individuals.
Collapse
Affiliation(s)
- Gang Wang
- School of Ulsan Ship and Ocean College, Ludong University, Yantai, China
| | - Wenju Zhou
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai, China
| | - Deping Kong
- School of Information and Electrical Engineering, Ludong University, Yantai, China
| | - Zongshuai Qu
- School of Information and Electrical Engineering, Ludong University, Yantai, China
| | - Maowen Ba
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Jinguang Hao
- School of Information and Electrical Engineering, Ludong University, Yantai, China
| | - Tao Yao
- School of Information and Electrical Engineering, Ludong University, Yantai, China
| | - Qunxi Dong
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, China
- School of Computing and Augmented Intelligence, Arizona State University, Tempe, AZ, USA
| | - Yi Su
- Banner Alzheimer’s Institute, 100 Washtenaw Avenue, Phoenix, AZ, USA
| | - Eric M Reiman
- Banner Alzheimer’s Institute, 100 Washtenaw Avenue, Phoenix, AZ, USA
| | | | - Kewei Chen
- Banner Alzheimer’s Institute, 100 Washtenaw Avenue, Phoenix, AZ, USA
| | - Yalin Wang
- School of Computing and Augmented Intelligence, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
34
|
Gao F. Integrated Positron Emission Tomography/Magnetic Resonance Imaging in clinical diagnosis of Alzheimer's disease. Eur J Radiol 2021; 145:110017. [PMID: 34826792 DOI: 10.1016/j.ejrad.2021.110017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/30/2021] [Accepted: 10/31/2021] [Indexed: 12/01/2022]
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disease which seriously endangers the health of the aged, is the most common etiology of senile dementia. With the increasing progress of neuroimaging technology, more and more imaging methods have been applied to study Alzheimer's disease. The emergence of integrated PET/MRI (Positron Emission Tomography/Magnetic Resonance Imaging) is a major advance in multimodal molecular imaging with many advantages on the structure of resolution and contrast of image over computed tomography (CT), PET and MRI. PET/MRI is now used stepwise in neurodegenerative diseases, and also has broad prospect of application in the early diagnosis of AD. In this review, we emphatically introduce the imaging advances of AD including functional imaging and molecular imaging, the advantages of PET/MRI over other imaging methods and prospects of PET/MRI in AD clinical diagnosis, especially in early diagnosis, clinical assessment and prediction on AD.
Collapse
Affiliation(s)
- Feng Gao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
35
|
Hin N, Newman M, Pederson S, Lardelli M. Iron Responsive Element-Mediated Responses to Iron Dyshomeostasis in Alzheimer's Disease. J Alzheimers Dis 2021; 84:1597-1630. [PMID: 34719489 DOI: 10.3233/jad-210200] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Iron trafficking and accumulation is associated with Alzheimer's disease (AD) pathogenesis. However, the role of iron dyshomeostasis in early disease stages is uncertain. Currently, gene expression changes indicative of iron dyshomeostasis are not well characterized, making it difficult to explore these in existing datasets. OBJECTIVE To identify sets of genes predicted to contain iron responsive elements (IREs) and use these to explore possible iron dyshomeostasis-associated gene expression responses in AD. METHODS Comprehensive sets of genes containing predicted IRE or IRE-like motifs in their 3' or 5' untranslated regions (UTRs) were identified in human, mouse, and zebrafish reference transcriptomes. Further analyses focusing on these genes were applied to a range of cultured cell, human, mouse, and zebrafish gene expression datasets. RESULTS IRE gene sets are sufficiently sensitive to distinguish not only between iron overload and deficiency in cultured cells, but also between AD and other pathological brain conditions. Notably, changes in IRE transcript abundance are among the earliest observable changes in zebrafish familial AD (fAD)-like brains, preceding other AD-typical pathologies such as inflammatory changes. Unexpectedly, while some IREs in the 3' untranslated regions of transcripts show significantly increased stability under iron deficiency in line with current assumptions, many such transcripts instead display decreased stability, indicating that this is not a generalizable paradigm. CONCLUSION Our results reveal IRE gene expression changes as early markers of the pathogenic process in fAD and are consistent with iron dyshomeostasis as an important driver of this disease. Our work demonstrates how differences in the stability of IRE-containing transcripts can be used to explore and compare iron dyshomeostasis-associated gene expression responses across different species, tissues, and conditions.
Collapse
Affiliation(s)
- Nhi Hin
- South Australian Genomics Centre, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA, Australia.,Alzheimer's Disease Genetics Laboratory, School of Biological Sciences, The University of Adelaide, North Terrace, Adelaide, SA, Australia
| | - Morgan Newman
- Alzheimer's Disease Genetics Laboratory, School of Biological Sciences, The University of Adelaide, North Terrace, Adelaide, SA, Australia
| | - Stephen Pederson
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, Faculty of Health & Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Michael Lardelli
- Alzheimer's Disease Genetics Laboratory, School of Biological Sciences, The University of Adelaide, North Terrace, Adelaide, SA, Australia
| |
Collapse
|
36
|
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: 52] [Impact Index Per Article: 13.0] [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
|
37
|
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: 123] [Impact Index Per Article: 30.8] [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
|
38
|
Strom A, Iaccarino L, Edwards L, Lesman-Segev OH, Soleimani-Meigooni DN, Pham J, Baker SL, Landau S, Jagust WJ, Miller BL, Rosen HJ, Gorno-Tempini ML, Rabinovici GD, La Joie R. Cortical hypometabolism reflects local atrophy and tau pathology in symptomatic Alzheimer's disease. Brain 2021; 145:713-728. [PMID: 34373896 PMCID: PMC9014741 DOI: 10.1093/brain/awab294] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 07/09/2021] [Accepted: 07/21/2021] [Indexed: 11/14/2022] Open
Abstract
Posterior cortical hypometabolism measured with [18F]-Fluorodeoxyglucose (FDG)-PET is a well-known marker of Alzheimer's disease-related neurodegeneration, but its associations with underlying neuropathological processes are unclear. We assessed cross-sectionally the relative contributions of three potential mechanisms causing hypometabolism in the retrosplenial and inferior parietal cortices: local molecular (amyloid and tau) pathology and atrophy, distant factors including contributions from the degenerating medial temporal lobe or molecular pathology in functionally connected regions, and the presence of the apolipoprotein E (APOE) ε4 allele. Two hundred and thirty-two amyloid-positive cognitively impaired patients from two cohorts (University of California, San Francisco, UCSF, and Alzheimer's Disease Neuroimaging Initiative, ADNI) underwent MRI and PET with FDG, amyloid-PET using [11C]-Pittsburgh Compound B, [18F]-Florbetapir, or [18F]-Florbetaben, and [18F]-Flortaucipir tau-PET within one year. Standard uptake value ratios (SUVR) were calculated using tracer-specific reference regions. Regression analyses were run within cohorts to identify variables associated with retrosplenial or inferior parietal FDG SUVR. On average, ADNI patients were older and were less impaired than UCSF patients. Regional patterns of hypometabolism were similar between cohorts, though there were cohort differences in regional gray matter atrophy. Local cortical thickness and tau-PET (but not amyloid-PET) were independently associated with both retrosplenial and inferior parietal FDG SUVR (ΔR2 = .09 to .21) across cohorts in models that also included age and disease severity (local model). Including medial temporal lobe volume improved the retrosplenial FDG model in ADNI (ΔR2 = .04, p = .008) but not UCSF (ΔR2 < .01, p = .52), and did not improve the inferior parietal models (ΔR2s < .01, ps > .37). Interaction analyses revealed that medial temporal volume was more strongly associated with retrosplenial FDG SUVR at earlier disease stages (p = .06 in UCSF, p = .046 in ADNI). Exploratory analyses across the cortex confirmed overall associations between hypometabolism and local tau pathology and thickness and revealed associations between medial temporal degeneration and hypometabolism in retrosplenial, orbitofrontal, and anterior cingulate cortices. Finally, our data did not support hypotheses of a detrimental effect of pathology in connected regions or of an effect of the APOE ε4 allele in impaired participants. Overall, in two independent groups of patients at symptomatic stages of Alzheimer's disease, cortical hypometabolism mainly reflected structural neurodegeneration and tau, but not amyloid, pathology.
Collapse
Affiliation(s)
- Amelia Strom
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Leonardo Iaccarino
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Lauren Edwards
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Orit H Lesman-Segev
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.,Department of Diagnostic Imaging, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - David N Soleimani-Meigooni
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Julie Pham
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Suzanne L Baker
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Susan Landau
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - William J Jagust
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.,Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Howard J Rosen
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Maria Luisa Gorno-Tempini
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.,Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.,Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA.,Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | | |
Collapse
|
39
|
Piersson AD, Mohamad M, Suppiah S, Rajab NF. Topographical patterns of whole-brain structural alterations in association with genetic risk, cerebrospinal fluid, positron emission tomography biomarkers of Alzheimer’s disease, and neuropsychological measures. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-021-00440-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
40
|
Knopman DS, Amieva H, Petersen RC, Chételat G, Holtzman DM, Hyman BT, Nixon RA, Jones DT. Alzheimer disease. Nat Rev Dis Primers 2021; 7:33. [PMID: 33986301 PMCID: PMC8574196 DOI: 10.1038/s41572-021-00269-y] [Citation(s) in RCA: 1185] [Impact Index Per Article: 296.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/09/2021] [Indexed: 12/21/2022]
Abstract
Alzheimer disease (AD) is biologically defined by the presence of β-amyloid-containing plaques and tau-containing neurofibrillary tangles. AD is a genetic and sporadic neurodegenerative disease that causes an amnestic cognitive impairment in its prototypical presentation and non-amnestic cognitive impairment in its less common variants. AD is a common cause of cognitive impairment acquired in midlife and late-life but its clinical impact is modified by other neurodegenerative and cerebrovascular conditions. This Primer conceives of AD biology as the brain disorder that results from a complex interplay of loss of synaptic homeostasis and dysfunction in the highly interrelated endosomal/lysosomal clearance pathways in which the precursors, aggregated species and post-translationally modified products of Aβ and tau play important roles. Therapeutic endeavours are still struggling to find targets within this framework that substantially change the clinical course in persons with AD.
Collapse
Affiliation(s)
| | - Helene Amieva
- Inserm U1219 Bordeaux Population Health Center, University of Bordeaux, Bordeaux, France
| | | | - Gäel Chételat
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, Cyceron, Caen, France
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Ralph A Nixon
- Departments of Psychiatry and Cell Biology, New York University Langone Medical Center, New York University, New York, NY, USA
- NYU Neuroscience Institute, New York University Langone Medical Center, New York University, New York, NY, USA
| | - David T Jones
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
41
|
Chen L, Wei Z, Chan KWY, Li Y, Suchal K, Bi S, Huang J, Xu X, Wong PC, Lu H, van Zijl PCM, Li T, Xu J. D-Glucose uptake and clearance in the tauopathy Alzheimer's disease mouse brain detected by on-resonance variable delay multiple pulse MRI. J Cereb Blood Flow Metab 2021; 41:1013-1025. [PMID: 32669023 PMCID: PMC8054725 DOI: 10.1177/0271678x20941264] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/29/2020] [Accepted: 06/17/2020] [Indexed: 12/11/2022]
Abstract
In this study, we applied on-resonance variable delay multiple pulse (onVDMP) MRI to study D-glucose uptake in a mouse model of Alzheimer's disease (AD) tauopathy and demonstrated its feasibility in discriminating AD mice from wild-type mice. The D-glucose uptake in the cortex of AD mice (1.70 ± 1.33%) was significantly reduced compared to that of wild-type mice (5.42 ± 0.70%, p = 0.0051). Also, a slower D-glucose uptake rate was found in the cerebrospinal fluid (CSF) of AD mice (0.08 ± 0.01 min-1) compared to their wild-type counterpart (0.56 ± 0.1 min-1, p < 0.001), which suggests the presence of an impaired glucose transporter on both blood-brain and blood-CSF barriers of these AD mice. Clearance of D-glucose was observed in the CSF of wild-type mice but not AD mice, which suggests dysfunction of the glymphatic system in the AD mice. The results in this study indicate that onVDMP MRI could be a cost-effective and widely available method for simultaneously evaluating glucose transporter and glymphatic function of AD. This study also suggests that tau protein affects the D-glucose uptake and glymphatic impairment in AD at a time point preceding neurofibrillary tangle pathology.
Collapse
Affiliation(s)
- Lin Chen
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhiliang Wei
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kannie WY Chan
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Yuguo Li
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kapil Suchal
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sheng Bi
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jianpan Huang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Xiang Xu
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Philip C Wong
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hanzhang Lu
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter CM van Zijl
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tong Li
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jiadi Xu
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
42
|
Zhang Q, Song Q, Gu X, Zheng M, Wang A, Jiang G, Huang M, Chen H, Qiu Y, Bo B, Tong S, Shao R, Li B, Wang G, Wang H, Hu Y, Chen H, Gao X. Multifunctional Nanostructure RAP-RL Rescues Alzheimer's Cognitive Deficits through Remodeling the Neurovascular Unit. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2001918. [PMID: 33511002 PMCID: PMC7816710 DOI: 10.1002/advs.202001918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/02/2020] [Indexed: 05/21/2023]
Abstract
Cerebrovascular dysfunction characterized by the neurovascular unit (NVU) impairment contributes to the pathogenesis of Alzheimer's disease (AD). In this study, a cerebrovascular-targeting multifunctional lipoprotein-biomimetic nanostructure (RAP-RL) constituted with an antagonist peptide (RAP) of receptor for advanced glycation end-products (RAGE), monosialotetrahexosyl ganglioside, and apolipoprotein E3 is developed to recover the functional NVU and normalize the cerebral vasculature. RAP-RL accumulates along the cerebral microvasculature through the specific binding of RAP to RAGE, which is overexpressed on cerebral endothelial cells in AD. It effectively accelerates the clearance of perivascular Aβ, normalizes the morphology and functions of cerebrovasculature, and restores the structural integrity and functions of NVU. RAP-RL markedly rescues the spatial learning and memory in APP/PS1 mice. Collectively, this study demonstrates the potential of the multifunctional nanostructure RAP-RL as a disease-modifying modality for AD treatment and provides the proof of concept that remodeling the functional NVU may represent a promising therapeutic approach toward effective intervention of AD.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Qingxiang Song
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Xiao Gu
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Mengna Zheng
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Antian Wang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Gan Jiang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Meng Huang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Huan Chen
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Yu Qiu
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Bin Bo
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong University800 Dongchuan RoadShanghai200240China
| | - Shanbao Tong
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong University800 Dongchuan RoadShanghai200240China
| | - Rong Shao
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Binyin Li
- Department of Neurology & Neuroscience InstituteRuijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine197 Rui Jin Er RoadShanghai200025China
| | - Gang Wang
- Department of Neurology & Neuroscience InstituteRuijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine197 Rui Jin Er RoadShanghai200025China
| | - Hao Wang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Yongbo Hu
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Hongzhuan Chen
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
- Institute of Interdisciplinary Integrative Biomedical ResearchShuguang HospitalShanghai University of Traditional Chinese Medicine1200 Cailun RoadShanghai201210China
| | - Xiaoling Gao
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| |
Collapse
|
43
|
Martí‐Juan G, Sanroma‐Guell G, Cacciaglia R, Falcon C, Operto G, Molinuevo JL, González Ballester MÁ, Gispert JD, Piella G, The Alzheimer's Disease Neuroimaging Initiative, The ALFA Study. Nonlinear interaction between APOE ε4 allele load and age in the hippocampal surface of cognitively intact individuals. Hum Brain Mapp 2021; 42:47-64. [PMID: 33017488 PMCID: PMC7721244 DOI: 10.1002/hbm.25202] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/16/2020] [Accepted: 08/11/2020] [Indexed: 01/27/2023] Open
Abstract
The ε4 allele of the gene Apolipoprotein E is the major genetic risk factor for Alzheimer's Disease. APOE ε4 has been associated with changes in brain structure in cognitively impaired and unimpaired subjects, including atrophy of the hippocampus, which is one of the brain structures that is early affected by AD. In this work we analyzed the impact of APOE ε4 gene dose and its association with age, on hippocampal shape assessed with multivariate surface analysis, in a ε4-enriched cohort of n = 479 cognitively healthy individuals. Furthermore, we sought to replicate our findings on an independent dataset of n = 969 individuals covering the entire AD spectrum. We segmented the hippocampus of the subjects with a multi-atlas-based approach, obtaining high-dimensional meshes that can be analyzed in a multivariate way. We analyzed the effects of different factors including APOE, sex, and age (in both cohorts) as well as clinical diagnosis on the local 3D hippocampal surface changes. We found specific regions on the hippocampal surface where the effect is modulated by significant APOE ε4 linear and quadratic interactions with age. We compared between APOE and diagnosis effects from both cohorts, finding similarities between APOE ε4 and AD effects on specific regions, and suggesting that age may modulate the effect of APOE ε4 and AD in a similar way.
Collapse
Affiliation(s)
- Gerard Martí‐Juan
- BCN MedTech, Departament de Tecnologies de la Informació i les ComunicacionsUniversitat Pompeu FabraBarcelonaSpain
| | | | - Raffaele Cacciaglia
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Carles Falcon
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Centro de Investigación Biomédica en Red de BioingenieríaBiomateriales y Nanomedicina (CIBERBBN)MadridSpain
| | - Grégory Operto
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
- Universitat Pompeu FabraBarcelonaSpain
| | - Miguel Ángel González Ballester
- BCN MedTech, Departament de Tecnologies de la Informació i les ComunicacionsUniversitat Pompeu FabraBarcelonaSpain
- ICREABarcelonaSpain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Centro de Investigación Biomédica en Red de BioingenieríaBiomateriales y Nanomedicina (CIBERBBN)MadridSpain
- Universitat Pompeu FabraBarcelonaSpain
| | - Gemma Piella
- BCN MedTech, Departament de Tecnologies de la Informació i les ComunicacionsUniversitat Pompeu FabraBarcelonaSpain
| | | | | |
Collapse
|
44
|
Mak E, Grigorova M, Beresford-Webb J, Malpetti M, Walpert M, Brown S, Jones E, Clare I, Hong YT, Fryer TD, Coles JP, Aigbirhio FI, Menon DK, Nestor PJ, Holland AJ, Zaman SH. Measuring cerebral perfusion with [ 11C]-PiB R1 in Down syndrome: associations with amyloid burden and longitudinal cognitive decline. Brain Commun 2020; 3:fcaa198. [PMID: 33543138 PMCID: PMC7849981 DOI: 10.1093/braincomms/fcaa198] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 08/11/2020] [Accepted: 08/31/2020] [Indexed: 11/14/2022] Open
Abstract
Positron emission tomography imaging of glucose hypometabolism and amyloid deposition are two well-established methods to evaluate preclinical changes in Alzheimer's disease and people with Down syndrome. However, the use of both imaging modalities may overburden participants, particularly those with intellectual disabilities and cognitive impairment. The relative tracer delivery of the [11C]-Pittsburgh Compound B has been proposed as a viable surrogate for cerebral perfusion. Here, we studied the impact of amyloid pathology on perfusion changes in Down syndrome and evaluated its associations with cognitive impairment. In total, 47 adults with Down syndrome underwent the [11C]-Pittsburgh Compound B imaging and structural imaging. The structural data were processed with Freesurfer to obtain anatomical segmentations and cortical thickness. The relative tracer delivery from [11C]-Pittsburgh Compound B was derived using a simplified reference tissue model. The sample was stratified into those with minimal amyloid burden (n = 25) and those with elevated amyloid (n = 22). We found significant and widespread reductions of cerebral perfusion in those with elevated amyloid burden, independent of age, gender, cognitive function and cortical thickness. In addition, cerebral perfusion was associated with the cognitive impairment among the Down syndrome group with elevated amyloid burden. These findings highlight the promising utility of the relative tracer delivery of the [11C]-Pittsburgh Compound B as a surrogate index in clinical trials for monitoring disease progression or tracking physiologic changes over time in Down syndrome.
Collapse
Affiliation(s)
- Elijah Mak
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge Biomedical Campus, CB2 0QQ, UK
| | - Monika Grigorova
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge Biomedical Campus, CB2 0QQ, UK
| | - Jessica Beresford-Webb
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge Biomedical Campus, CB2 0QQ, UK
| | - Maura Malpetti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - Madeline Walpert
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge Biomedical Campus, CB2 0QQ, UK
| | - Stephanie Brown
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge Biomedical Campus, CB2 0QQ, UK
| | - Elizabeth Jones
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge Biomedical Campus, CB2 0QQ, UK
| | - Isabel Clare
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge Biomedical Campus, CB2 0QQ, UK
| | - Young T Hong
- Wolfson Brain Imaging Centre, University of Cambridge, CB2 0QQ, UK
| | - Tim D Fryer
- Wolfson Brain Imaging Centre, University of Cambridge, CB2 0QQ, UK
| | - Jonathan P Coles
- Division of Anaesthesia, University of Cambridge, Cambridge, CB2 0QQ, UK
| | | | - David K Menon
- Division of Anaesthesia, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Peter J Nestor
- Queensland Brain Institute, University of Queensland, Queensland, QLD 4072, Australia
| | - Anthony J Holland
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge Biomedical Campus, CB2 0QQ, UK
| | - Shahid H Zaman
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge Biomedical Campus, CB2 0QQ, UK
| |
Collapse
|
45
|
Clinical Utility of the Pathogenesis-Related Proteins in Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21228661. [PMID: 33212853 PMCID: PMC7698353 DOI: 10.3390/ijms21228661] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/05/2020] [Accepted: 11/13/2020] [Indexed: 12/16/2022] Open
Abstract
Research on the Aβ cascade and alternations of biomarkers in neuro-inflammation, synaptic dysfunction, and neuronal injury followed by Aβ have progressed. But the question is how to use the biomarkers. Here, we examine the evidence and pathogenic implications of protein interactions and the time order of alternation. After the deposition of Aβ, the change of tau, neurofilament light chain (NFL), and neurogranin (Ng) is the main alternation and connection to others. Neuro-inflammation, synaptic dysfunction, and neuronal injury function is exhibited prior to the structural and metabolic changes in the brain following Aβ deposition. The time order of such biomarkers compared to the tau protein is not clear. Despite the close relationship between biomarkers and plaque Aβ deposition, several factors favor one or the other. There is an interaction between some proteins that can predict the brain amyloid burden. The Aβ cascade hypothesis could be the pathway, but not all subjects suffer from Alzheimer's disease (AD) within a long follow-up, even with very elevated Aβ. The interaction of biomarkers and the time order of change require further research to identify the right subjects and right molecular target for precision medicine therapies.
Collapse
|
46
|
Kulkarni P, Grant S, Morrison TR, Cai X, Iriah S, Kristal BS, Honeycutt J, Brenhouse H, Hartner JC, Madularu D, Ferris CF. Characterizing the human APOE epsilon 4 knock-in transgene in female and male rats with multimodal magnetic resonance imaging. Brain Res 2020; 1747:147030. [PMID: 32745658 DOI: 10.1016/j.brainres.2020.147030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/23/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
The APOE Ɛ4 genotype is the most prevalent genetic risk for Alzheimer's disease (AD). Women carriers of Ɛ4 have higher risk for an early onset of AD than men. Human imaging studies suggest apolipoprotein Ɛ4 may affect brain structures associated with cognitive decline in AD many years before disease onset. It was hypothesized that female APOE Ɛ4 carriers would present with decreased cognitive function and neuroradiological evidence of early changes in brain structure and function as compared to male carriers. Six-month old wild-type (WT) and human APOE Ɛ4 knock-in (TGRA8960), male and female Sprague Dawley rats were studied for changes in brain structure using voxel-based morphometry, alteration in white and gray matter microarchitecture using diffusion weighted imaging with indices of anisotropy, and functional coupling using resting state BOLD functional connectivity. Images from each modality were registered to, and analyzed, using a 3D MRI rat atlas providing site-specific data on over 168 different brain areas. Quantitative volumetric analysis revealed areas involved in memory and arousal were significantly different between Ɛ4 and wild-type (WT) females, with few differences between male genotypes. Diffusion weighted imaging showed few differences between WT and Ɛ4 females, while male genotypes showed significant different measures in fractional anisotropy and apparent diffusion coefficient. Resting state functional connectivity showed Ɛ4 females had greater connectivity between areas involved in cognition, emotion, and arousal compared to WT females, with male Ɛ4 showing few differences from controls. Interestingly, male Ɛ4 showed increased anxiety and decreased performance in spatial and episodic memory tasks compared to WT males, with female genotypes showing little difference across behavioral tests. The sex differences in behavior and diffusion weighted imaging suggest male carriers of the Ɛ4 allele may be more vulnerable to cognitive and emotional complications compared to female carriers early in life. Conversely, the data may also suggest that female carriers are more resilient to cognitive/emotional problems at this stage of life perhaps due to altered brain volumes and enhanced connectivity.
Collapse
Affiliation(s)
- Praveen Kulkarni
- Northeastern Univ, Center for Translational NeuroImaging, Boston, MA, United States
| | - Simone Grant
- Dept of Psychiatry and Neurosciences, Univ California at Davis, United States
| | - Thomas R Morrison
- Northeastern Univ, Center for Translational NeuroImaging, Boston, MA, United States
| | - Xuezhu Cai
- Northeastern Univ, Center for Translational NeuroImaging, Boston, MA, United States
| | - Sade Iriah
- Northeastern Univ, Center for Translational NeuroImaging, Boston, MA, United States
| | - Bruce S Kristal
- Dept Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | | | | | | | - Dan Madularu
- Northeastern Univ, Center for Translational NeuroImaging, Boston, MA, United States
| | - Craig F Ferris
- Northeastern Univ, Center for Translational NeuroImaging, Boston, MA, United States; Northeastern Univ, Dept. Pharmaceutical Sciences, Boston, MA, United States.
| |
Collapse
|
47
|
Malykhin NV, Travis S, Fujiwara E, Huang Y, Camicioli R, Olsen F. The associations of the
BDNF
and
APOE
polymorphisms, hippocampal subfield volumes, and episodic memory performance across the lifespan. Hippocampus 2020; 30:1081-1097. [DOI: 10.1002/hipo.23217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 03/23/2020] [Accepted: 04/25/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Nikolai V. Malykhin
- Neuroscience and Mental Health Institute University of Alberta Edmonton Alberta Canada
- Department of Biomedical Engineering University of Alberta Edmonton Alberta Canada
- Department of Psychiatry University of Alberta Edmonton Alberta Canada
| | - Scott Travis
- Neuroscience and Mental Health Institute University of Alberta Edmonton Alberta Canada
| | - Esther Fujiwara
- Neuroscience and Mental Health Institute University of Alberta Edmonton Alberta Canada
- Department of Psychiatry University of Alberta Edmonton Alberta Canada
| | - Yushan Huang
- Department of Biomedical Engineering University of Alberta Edmonton Alberta Canada
| | | | - Fraser Olsen
- Department of Biomedical Engineering University of Alberta Edmonton Alberta Canada
| |
Collapse
|
48
|
Gaitán JM, Boots EA, Dougherty RJ, Ma Y, Edwards DF, Mitchell CC, Christian BT, Cook DB, Okonkwo OC. Protocol of Aerobic Exercise and Cognitive Health (REACH): A Pilot Study. J Alzheimers Dis Rep 2020; 4:107-121. [PMID: 32587945 PMCID: PMC7306918 DOI: 10.3233/adr-200180] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2020] [Indexed: 12/29/2022] Open
Abstract
A growing body of evidence supports that aerobic exercise can decrease the risk of future cognitive impairment and Alzheimer's disease (AD). There is a pressing need to rigorously determine whether cognitively normal yet at-risk individuals stand to benefit from the protective effects of exercise. The present study will test the feasibility of an aerobic exercise intervention in such a population and inform the design of a larger-scale randomized, controlled trial examining the effect of aerobic exercise on biomarkers of AD in late-middle-aged, at-risk individuals. This was a single-site, 1 : 1 block-randomized, parallel, two-arm trial. Cognitively normal participants aged 45-80 with documentation of familial and genetic AD risk factors were randomly assigned to one of two interventions. The Usual Physical Activity group was provided educational materials about exercise. The Enhanced Physical Activity intervention delivered 26 weeks of individualized and supervised aerobic exercise. Exercise duration and intensity were incrementally increased to 150 min/week and 70-80% of heart rate reserve, respectively. Retention and adherence were measured to assess study feasibility. In addition, pre- and post- intervention differences between the two arms were evaluated for cardiorespiratory fitness, physical activity, brain glucose metabolism, cerebral structure, vascular health, memory, executive function, and mood. Data from randomized controlled trials of exercise training are needed to identify the proper exercise prescription for reducing accumulation of AD biomarkers in cognitively normal individuals. The current trial will contribute to filling that gap while informing the design of large-scale trials.
Collapse
Affiliation(s)
- Julian M. Gaitán
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Elizabeth A. Boots
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Ryan J. Dougherty
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Kinesiology, University of Wisconsin School of Education, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Yue Ma
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Dorothy F. Edwards
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Carol C. Mitchell
- Department of Medicine, Cardiovascular Medicine Division, University of Wisconsin Atherosclerosis Imaging Research Program, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Bradley T. Christian
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Dane B. Cook
- Department of Kinesiology, University of Wisconsin School of Education, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Ozioma C. Okonkwo
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
49
|
Volumetric alterations in the hippocampal subfields of subjects at increased risk of dementia. Neurobiol Aging 2020; 91:36-44. [PMID: 32311609 DOI: 10.1016/j.neurobiolaging.2020.03.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/04/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022]
Abstract
The hippocampus is one of the first regions to demonstrate atrophy during the prodromal stage of Alzheimer's disease. Volumetric analysis of its individual subfields could provide biomarkers with higher sensitivity than whole hippocampal volume during an earlier disease stage. We quantified the hippocampal subfields volume in a large cohort comprising healthy participants (aged 40-59) with dementia family history (FH) and controls (without FH), examined at 2 time points across 2 years. Subfield volumes were quantified using both a T1-weighted and a high-resolution T2 hippocampal magnetic resonance imaging acquisition with Freesurfer. The participants were stratified based on dementia FH, APOE genotype, and CAIDE (Cardiovascular Risk Factors, Aging and Dementia) risk score. Whole hippocampal volume did not differ between the groups. The volume of the molecular layer was lower in participants with an APOE ε4 genotype, but there were no differences between subjects with and without dementia FH or with an increasing CAIDE score. The molecular layer may be the first hippocampal region to demonstrate volumetric alterations in subjects at risk of dementia.
Collapse
|
50
|
Impairment of Glycolysis-Derived l-Serine Production in Astrocytes Contributes to Cognitive Deficits in Alzheimer's Disease. Cell Metab 2020; 31:503-517.e8. [PMID: 32130882 DOI: 10.1016/j.cmet.2020.02.004] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/25/2019] [Accepted: 02/07/2020] [Indexed: 12/11/2022]
Abstract
Alteration of brain aerobic glycolysis is often observed early in the course of Alzheimer's disease (AD). Whether and how such metabolic dysregulation contributes to both synaptic plasticity and behavioral deficits in AD is not known. Here, we show that the astrocytic l-serine biosynthesis pathway, which branches from glycolysis, is impaired in young AD mice and in AD patients. l-serine is the precursor of d-serine, a co-agonist of synaptic NMDA receptors (NMDARs) required for synaptic plasticity. Accordingly, AD mice display a lower occupancy of the NMDAR co-agonist site as well as synaptic and behavioral deficits. Similar deficits are observed following inactivation of the l-serine synthetic pathway in hippocampal astrocytes, supporting the key role of astrocytic l-serine. Supplementation with l-serine in the diet prevents both synaptic and behavioral deficits in AD mice. Our findings reveal that astrocytic glycolysis controls cognitive functions and suggest oral l-serine as a ready-to-use therapy for AD.
Collapse
|