1
|
Gomes Gonçalves N, Cacau LT, Ferreira NV, Lotufo PA, Goulart AC, Viana MC, Barreto SM, Bensenor IM, Marchioni DM, Suemoto CK. Adherence to the planetary health diet and cognitive decline: findings from the ELSA-Brasil study. NATURE AGING 2024; 4:1465-1476. [PMID: 38942982 DOI: 10.1038/s43587-024-00666-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 06/13/2024] [Indexed: 06/30/2024]
Abstract
The EAT-Lancet Commission proposed a planetary health diet to improve human health within planetary boundaries; however, little is known about the association between adherence to this diet and cognitive decline. We used data from three waves of the Brazilian Longitudinal Study of Adult Health to evaluate the association between the planetary health diet and cognitive decline using linear mixed-effects models. Here we show that in 11,737 participants (mean (s.d.) age 51.6 (9.0) years, 54% women and 53% white), higher adherence to the planetary health diet was associated with slower memory decline (P = 0.046) and that income was a modifier in this association (P < 0.001). Adherence to the planetary health diet was associated with slower decline of memory (P = 0.040) and global cognition (P = 0.009) in high-income participants. No association was found among low-income participants. The results of our study highlight that the promotion of healthy dietary patterns should take into consideration income barriers as well as differences in dietary habits to achieve high adherence.
Collapse
Affiliation(s)
| | - Leandro Teixeira Cacau
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | | | - Paulo Andrade Lotufo
- Center for Clinical and Epidemiological Research, Hospital Universitario, University of São Paulo, São Paulo, Brazil
| | - Alessandra Carvalho Goulart
- Center for Clinical and Epidemiological Research, Hospital Universitario, University of São Paulo, São Paulo, Brazil
| | - Maria Carmen Viana
- Department of Social Medicine, Federal University of Espírito Santo, Vitória, Brazil
| | - Sandhi Maria Barreto
- Preventive and Social Medicine Department, Clinical Hospital/ EBSERH, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Isabela Martins Bensenor
- Center for Clinical and Epidemiological Research, Hospital Universitario, University of São Paulo, São Paulo, Brazil
| | - Dirce Maria Marchioni
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
2
|
Norambuena A, Sagar VK, Wang Z, Raut P, Feng Z, Wallrabe H, Pardo E, Kim T, Alam SR, Hu S, Periasamy A, Bloom GS. Disrupted mitochondrial response to nutrients is a presymptomatic event in the cortex of the APP SAA knock-in mouse model of Alzheimer's disease. Alzheimers Dement 2024; 20:6844-6859. [PMID: 39171353 PMCID: PMC11485302 DOI: 10.1002/alz.14144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/13/2024] [Accepted: 07/01/2024] [Indexed: 08/23/2024]
Abstract
INTRODUCTION Reduced brain energy metabolism, mammalian target of rapamycin (mTOR) dysregulation, and extracellular amyloid beta (Aβ) oligomer (xcAβO) buildup are some well-known Alzheimer's disease (AD) features; how they promote neurodegeneration is poorly understood. We previously reported that xcAβOs inhibit nutrient-induced mitochondrial activity (NiMA) in cultured neurons. We now report NiMA disruption in vivo. METHODS Brain energy metabolism and oxygen consumption were recorded in heterozygous amyloid precursor protein knock-in (APPSAA) mice using two-photon fluorescence lifetime imaging and multiparametric photoacoustic microscopy. RESULTS NiMA is inhibited in APPSAA mice before other defects are detected in these Aβ-producing animals that do not overexpress APP or contain foreign DNA inserts into genomic DNA. Glycogen synthase kinase 3 (GSK3β) signals through mTORC1 to regulate NiMA independently of mitochondrial biogenesis. Inhibition of GSK3β with TWS119 stimulates NiMA in cultured human neurons, and mitochondrial activity and oxygen consumption in APPSAA mice. DISCUSSION NiMA disruption in vivo occurs before plaques, neuroinflammation, and cognitive decline in APPSAA mice, and may represent an early stage in human AD. HIGHLIGHTS Amyloid beta blocks communication between lysosomes and mitochondria in vivo. Nutrient-induced mitochondrial activity (NiMA) is disrupted long before the appearance of Alzheimer's disease (AD) histopathology in heterozygous amyloid precursor protein knock-in (APPSAA/+) mice. NiMA is disrupted long before learning and memory deficits in APPSAA/+ mice. Pharmacological interventions can rescue AD-related NiMA disruption in vivo.
Collapse
Affiliation(s)
- Andrés Norambuena
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Vijay Kumar Sagar
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- W.M. Keck Center for Cellular ImagingUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Zhuoying Wang
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Prakash Raut
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Ziang Feng
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Horst Wallrabe
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- W.M. Keck Center for Cellular ImagingUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Evelyn Pardo
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Taylor Kim
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Shagufta Rehman Alam
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- W.M. Keck Center for Cellular ImagingUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Song Hu
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Ammasi Periasamy
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- W.M. Keck Center for Cellular ImagingUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - George S. Bloom
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Cell BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of NeuroscienceUniversity of VirginiaCharlottesvilleVirginiaUSA
| |
Collapse
|
3
|
Jin S, Wang J, He Y. The brain network hub degeneration in Alzheimer's disease. BIOPHYSICS REPORTS 2024; 10:213-229. [PMID: 39281195 PMCID: PMC11399886 DOI: 10.52601/bpr.2024.230025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 04/26/2024] [Indexed: 09/18/2024] Open
Abstract
Alzheimer's disease (AD) has been conceptualized as a syndrome of brain network dysfunction. Recent imaging connectomics studies have provided unprecedented opportunities to map structural and functional brain networks in AD. By reviewing molecular, imaging, and computational modeling studies, we have shown that highly connected brain hubs are primarily distributed in the medial and lateral prefrontal, parietal, and temporal regions in healthy individuals and that the hubs are selectively and severely affected in AD as manifested by increased amyloid-beta deposition and regional atrophy, hypo-metabolism, and connectivity dysfunction. Furthermore, AD-related hub degeneration depends on the imaging modality with the most notable degeneration in the medial temporal hubs for morphological covariance networks, the prefrontal hubs for structural white matter networks, and in the medial parietal hubs for functional networks. Finally, the AD-related hub degeneration shows metabolic, molecular, and genetic correlates. Collectively, we conclude that the brain-network-hub-degeneration framework is promising to elucidate the biological mechanisms of network dysfunction in AD, which provides valuable information on potential diagnostic biomarkers and promising therapeutic targets for the disease.
Collapse
Affiliation(s)
- Suhui Jin
- Institute for Brain Research and Rehabilitation, Guangzhou 510631, China
| | - Jinhui Wang
- Institute for Brain Research and Rehabilitation, Guangzhou 510631, China
- Guangdong Key Laboratory of Mental Health and Cognitive Science, Guangzhou 510631, China
- Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Yong He
- IDG/McGovern Institute for Brain Research, Beijing 100875, China
- National Key Laboratory of Cognitive Neuroscience and Learning, Beijing 100875, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
4
|
Ebright B, Duro MV, Chen K, Louie S, Yassine HN. Effects of APOE4 on omega-3 brain metabolism across the lifespan. Trends Endocrinol Metab 2024; 35:745-757. [PMID: 38609814 PMCID: PMC11321946 DOI: 10.1016/j.tem.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 03/09/2024] [Accepted: 03/14/2024] [Indexed: 04/14/2024]
Abstract
Omega-3 (n-3) polyunsaturated fatty acids (PUFAs), such as docosahexaenoic acid (DHA), have important roles in human nutrition and brain health by promoting neuronal functions, maintaining inflammatory homeostasis, and providing structural integrity. As Alzheimer's disease (AD) pathology progresses, DHA metabolism in the brain becomes dysregulated, the timing and extent of which may be influenced by the apolipoprotein E ε4 (APOE4) allele. Here, we discuss how maintaining adequate DHA intake early in life may slow the progression to AD dementia in cognitively normal individuals with APOE4, how recent advances in DHA brain imaging could offer insights leading to more personalized preventive strategies, and how alternative strategies targeting PUFA metabolism pathways may be more effective in mitigating disease progression in patients with existing AD dementia.
Collapse
Affiliation(s)
- Brandon Ebright
- Department of Clinical Pharmacy, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Marlon V Duro
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Kai Chen
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Stan Louie
- Department of Clinical Pharmacy, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Hussein N Yassine
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
5
|
Lázaro I, Grau‐Rivera O, Suárez‐Calvet M, Fauria K, Minguillón C, Shekari M, Falcón C, García‐Prat M, Huguet J, Molinuevo JL, Gispert J, Sala‐Vila A. Omega-3 blood biomarkers relate to brain glucose uptake in individuals at risk of Alzheimer's disease dementia. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12596. [PMID: 38974876 PMCID: PMC11224768 DOI: 10.1002/dad2.12596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/26/2024] [Accepted: 03/13/2024] [Indexed: 07/09/2024]
Abstract
INTRODUCTION Brain glucose hypometabolism is a preclinical feature of Alzheimer's disease (AD). Dietary omega-3 fatty acids promote brain glucose metabolism, but clinical research is incipient. Circulating omega-3s objectively reflect their dietary intake. METHODS This was a cross-sectional study in 320 cognitively unimpaired participants at increased risk of AD dementia. Using lipidomics, we determined blood docosahexaenoic (DHA) and alpha-linolenic (ALA) acid levels (omega-3s from marine and plant origin, respectively). We assessed brain glucose metabolism using [18-F]-fluorodeoxyglucose (FDG) positron emission tomography (PET). RESULTS Blood ALA directly related to FDG uptake in brain areas known to be affected in AD. Stronger associations were observed in apolipoprotein E ε4 carriers and homozygotes. For DHA, significant direct associations were restricted to amyloid beta-positive tau-positive participants. DISCUSSION Blood omega-3 directly relate to preserved glucose metabolism in AD-vulnerable brain regions in individuals at increased risk of AD dementia. This adds to the benefits of omega-3 supplementation in the preclinical stage of AD dementia. Highlights Blood omega-3s were related to brain glucose uptake in participants at risk of Alzheimer's disease (AD) dementia.Complementary associations were observed for omega-3 from marine and plant sources.Foods rich in omega-3 might be useful in early features of AD.
Collapse
Affiliation(s)
- Iolanda Lázaro
- Hospital del Mar Research InstituteBarcelonaSpain
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN)Instituto de Salud Carlos IIIMadridSpain
| | - Oriol Grau‐Rivera
- Hospital del Mar Research InstituteBarcelonaSpain
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)Instituto de Salud Carlos IIIMadridSpain
- Servei de NeurologiaHospital del MarBarcelonaSpain
| | - Marc Suárez‐Calvet
- Hospital del Mar Research InstituteBarcelonaSpain
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)Instituto de Salud Carlos IIIMadridSpain
- Servei de NeurologiaHospital del MarBarcelonaSpain
| | - Karine Fauria
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)Instituto de Salud Carlos IIIMadridSpain
| | - Carolina Minguillón
- Hospital del Mar Research InstituteBarcelonaSpain
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)Instituto de Salud Carlos IIIMadridSpain
| | - Mahnaz Shekari
- Hospital del Mar Research InstituteBarcelonaSpain
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)Instituto de Salud Carlos IIIMadridSpain
- Department of Medicine and Life SciencesUniversitat Pompeu FabraBarcelonaSpain
| | - Carles Falcón
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red BioingenieríaBiomateriales y Nanomedicina (CIBERBBN)Instituto de Salud Carlos IIIMadridSpain
| | - Marina García‐Prat
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
| | - Jordi Huguet
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Lundbeck A/SCopenhagenDenmark
| | - Juan‐Domingo Gispert
- Hospital del Mar Research InstituteBarcelonaSpain
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red BioingenieríaBiomateriales y Nanomedicina (CIBERBBN)Instituto de Salud Carlos IIIMadridSpain
| | - Aleix Sala‐Vila
- Hospital del Mar Research InstituteBarcelonaSpain
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN)Instituto de Salud Carlos IIIMadridSpain
- Fatty Acid Research InstituteSioux FallsSouth DakotaUSA
| | | |
Collapse
|
6
|
Thomas A, Ryan CP, Caspi A, Liu Z, Moffitt TE, Sugden K, Zhou J, Belsky DW, Gu Y. Diet, Pace of Biological Aging, and Risk of Dementia in the Framingham Heart Study. Ann Neurol 2024; 95:1069-1079. [PMID: 38407506 PMCID: PMC11102315 DOI: 10.1002/ana.26900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 01/16/2024] [Accepted: 02/05/2024] [Indexed: 02/27/2024]
Abstract
OBJECTIVE People who eat healthier diets are less likely to develop dementia, but the biological mechanism of this protection is not well understood. We tested the hypothesis that healthy diet protects against dementia because it slows the pace of biological aging. METHODS We analyzed Framingham Offspring Cohort data. We included participants ≥60 years-old, free of dementia and having dietary, epigenetic, and follow-up data. We assessed healthy diet as long-term adherence to the Mediterranean-Dash Intervention for Neurodegenerative Delay diet (MIND, over 4 visits spanning 1991-2008). We measured the pace of aging from blood DNA methylation data collected in 2005-2008 using the DunedinPACE epigenetic clock. Incident dementia and mortality were defined using study records compiled from 2005 to 2008 visit through 2018. RESULTS Of n = 1,644 included participants (mean age 69.6, 54% female), n = 140 developed dementia and n = 471 died over 14 years of follow-up. Greater MIND score was associated with slower DunedinPACE and reduced risks for dementia and mortality. Slower DunedinPACE was associated with reduced risks for dementia and mortality. In mediation analysis, slower DunedinPACE accounted for 27% of the diet-dementia association and 57% of the diet-mortality association. INTERPRETATION Findings suggest that slower pace of aging mediates part of the relationship of healthy diet with reduced dementia risk. Monitoring pace of aging may inform dementia prevention. However, a large fraction of the diet-dementia association remains unexplained and may reflect direct connections between diet and brain aging that do not overlap other organ systems. Investigation of brain-specific mechanisms in well-designed mediation studies is warranted. ANN NEUROL 2024;95:1069-1079.
Collapse
Affiliation(s)
- Aline Thomas
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
- Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Calen P. Ryan
- Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| | - Avshalom Caspi
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
| | - Zhonghua Liu
- Department of Biostatistics, Columbia University, New York, NY 10032, USA
| | - Terrie E. Moffitt
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
| | - Karen Sugden
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
| | - Jiayi Zhou
- Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| | - Daniel W. Belsky
- Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY 10032, USA
- Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| | - Yian Gu
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
- Department of Neurology, Columbia University, New York, NY 10032, USA
- Department of Biostatistics, Columbia University, New York, NY 10032, USA
- Gertrude H. Sergievsky Center, Columbia University, New York, NY 10032, USA
| |
Collapse
|
7
|
Hernandez AR, Parker E, Babar M, Banerjee A, Ding S, Simley A, Buford TW. Microbiome-driven alterations in metabolic pathways and impaired cognition in aged female TgF344-AD rats. AGING BRAIN 2024; 5:100119. [PMID: 38881651 PMCID: PMC11179252 DOI: 10.1016/j.nbas.2024.100119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/09/2024] [Accepted: 05/28/2024] [Indexed: 06/18/2024] Open
Abstract
Alzheimer's disease (AD) not only affects cognition and neuropathology, but several other facets capable of negatively impacting quality of life and potentially driving impairments, including altered gut microbiome (GMB) composition and metabolism. Aged (20 + mo) female TgF344-AD and wildtype rats were cognitively characterized on several tasks incorporating several cognitive domains, including task acquisition, object recognition memory, anxiety-like behaviors, and spatial navigation. Additionally, metabolic phenotyping, GMB sequencing throughout the intestinal tract (duodenum, jejunum, ileum, colon, and feces), neuropathological burden assessment and marker gene functional abundance predictions (PICRUSt2) were conducted. TgF344-AD rats demonstrated significant cognitive impairment in multiple domains, as well as regionally specific GMB dysbiosis. Relationships between peripheral factors were investigated using Canonical Correspondence Analysis (CCA), revealing correlations between GMB changes and both cognitive and metabolic factors. Moreover, communities of gut microbes contributing to essential metabolic pathways were significantly altered in TgF344-AD rats. These data indicate dysbiosis may affect cognitive outcomes in AD through alterations in metabolism-related enzymatic pathways that are necessary for proper brain function. Moreover, these changes were mostly observed in intestinal segments required for carbohydrate digestion, not fecal samples. These data support the targeting of intestinal and microbiome health for the treatment of AD.
Collapse
Affiliation(s)
- Abbi R Hernandez
- Department of Medicine, Division of Geriatrics, Gerontology & Palliative Care, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Erik Parker
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University-Bloomington, Bloomington, IN 47405, USA
| | - Maham Babar
- Department of Medicine, Division of Geriatrics, Gerontology & Palliative Care, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Anisha Banerjee
- Department of Medicine, Division of Geriatrics, Gerontology & Palliative Care, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Sarah Ding
- Department of Medicine, Division of Geriatrics, Gerontology & Palliative Care, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Alexis Simley
- Department of Medicine, Division of Geriatrics, Gerontology & Palliative Care, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Thomas W Buford
- Department of Medicine, Division of Geriatrics, Gerontology & Palliative Care, University of Alabama at Birmingham, Birmingham, AL 35205, USA
- Birmingham/Atlanta VA GRECC, Birmingham VA Medical Center, Birmingham, AL 35244, USA
| |
Collapse
|
8
|
Alam J, Kalash A, Hassan MI, Rahman SZ. Agents at the Peak of US FDA Approval for the Treatment of Alzheimer's Disease. Neurol Res 2024; 46:318-325. [PMID: 38197595 DOI: 10.1080/01616412.2024.2302271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024]
Abstract
Where Alzheimer's disease (AD) is becoming a global health issue, the present anti-AD medications have also been exposed to produce only symptomatic outcomes. The pathological factors, like neuronal transmission impairment, amyloidal-tau constituents, oxidative damage, neuro-inflammation, synaptic dysfunction, infectious agents, and impairment of gut microbiota and vitamins' levels; all favor the disease's progression and sustainability. The researchers have investigated several drugable molecules against these factors; however, no treatment could have been discovered yet to prevent the disease's progression rather than anti-amyloidal antibodies. After a comprehensive review of the literature and the clinical registry (clinicaltrials.gov), the authors of this manuscript have explored drug molecules that are under phase-3 of clinical trials and at the peak of getting approval for the management of AD. The inclusion and exclusion criteria for clinical trials were decided by considering the basis of a drug's approval. We included only the clinical trials were found in stages of Enrolling-by-Invitation, Recruiting, Not Recruiting (But active), and Not Recruiting (Not active) while excluding Completed, Terminated, Suspended, Withdrawn, or the trials of Unknown Status. We have found many potent drug molecules reached the clinical trials in phase-3 that could be futuristic anti-AD agents. This review article aims to provide an update on the prospective potential anti-AD medicines and to reveal the therapeutic targets of great significance for designing further a possible drug development strategy against AD pathology.
Collapse
Affiliation(s)
- Jahngeer Alam
- Department of Pharmacology, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Anushka Kalash
- Department of Pharmacology, School of Pharmaceutical Sciences, Jaipur National University, Jaipur, Rajasthan, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Syed Ziaur Rahman
- Department of Pharmacology, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| |
Collapse
|
9
|
Buchholz A, Deme P, Betz JF, Brandt J, Haughey N, Cervenka MC. A randomized feasibility trial of the modified Atkins diet in older adults with mild cognitive impairment due to Alzheimer's disease. Front Endocrinol (Lausanne) 2024; 15:1182519. [PMID: 38505743 PMCID: PMC10949529 DOI: 10.3389/fendo.2024.1182519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 02/05/2024] [Indexed: 03/21/2024] Open
Abstract
Background Alzheimer's disease (AD) is increasing in prevalence, but effective treatments for its cognitive impairment remain severely limited. This study investigates the impact of ketone body production through dietary manipulation on memory in persons with mild cognitive impairment due to early AD and explores potential mechanisms of action. Methods We conducted a 12-week, parallel-group, controlled feasibility trial of a ketogenic diet, the modified Atkins diet (MAD), compared to a control diet in patients with cognitive impairments attributed to AD. We administered neuropsychological assessments, including memory tests, and collected blood samples at baseline and after 12 weeks of intervention. We performed untargeted lipidomic and targeted metabolomic analyses on plasma samples to detect changes over time. Results A total of 839 individuals were screened to yield 38 randomized participants, with 20 assigned to receive MAD and 18 assigned to receive a control diet. Due to attrition, only 13 in the MAD arm and nine in the control arm were assessed for the primary endpoint, with two participants meeting ketosis levels used to define MAD adherence criteria. The average change from baseline in the Memory Composite Score was 1.37 (95% CI: -0.87, 4.90) points higher in the MAD group compared to the control group. The effect size of the intervention on baseline MAD change was moderate (Cohen's D = 0.57, 95% CI: -0.67, 1.33). In the 15 participants (nine MAD, six control) assessed for lipidomic and metabolomic-lipidomics and metabolomics, 13 metabolites and 10 lipids showed significant changes from baseline to 12 weeks, including triacylglycerols (TAGs, 50:5, 52:5, and 52:6), sphingomyelins (SM, 44:3, 46:0, 46:3, and 48:1), acetoacetate, fatty acylcarnitines, glycerol-3-phosphate, and hydroxy fatty acids. Conclusions Attrition was greatest between baseline and week 6. All participants retained at week 6 completed the study. Despite low rates of adherence by criteria defined a priori, lipidomic and metabolomic analyses indicate significant changes from baseline in circulating lipids and metabolites between MAD and control participants at 12-week postrandomization, and MAD participants showed greater, albeit nonsignificant, improvement in memory.
Collapse
Affiliation(s)
- Alison Buchholz
- Department of Psychiatry & Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Pragney Deme
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Joshua F. Betz
- Department of Biostatistics, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Jason Brandt
- Department of Psychiatry & Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Norman Haughey
- Department of Psychiatry & Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mackenzie C. Cervenka
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
10
|
Norambuena A, Sagar VK, Wang Z, Raut P, Feng Z, Wallrabe H, Pardo E, Kim T, Alam SR, Hu S, Periasamy A, Bloom GS. Disrupted mitochondrial response to nutrients is a presymptomatic event in the cortex of the APP SAA knock-in mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.02.578668. [PMID: 38352486 PMCID: PMC10862844 DOI: 10.1101/2024.02.02.578668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Introduction Reduced brain energy metabolism, mTOR dysregulation, and extracellular amyloid-β oligomer (xcAβO) buildup characterize AD; how they collectively promote neurodegeneration is poorly understood. We previously reported that xcAβOs inhibit N utrient-induced M itochondrial A ctivity (NiMA) in cultured neurons. We now report NiMA disruption in vivo . Methods Brain energy metabolism and oxygen consumption were recorded in APP SAA/+ mice using two-photon fluorescence lifetime imaging and multiparametric photoacoustic microscopy. Results NiMA is inhibited in APP SAA/+ mice before other defects are detected in these amyloid-β-producing animals that do not overexpress APP or contain foreign DNA inserts into genomic DNA. GSK3β signals through mTORC1 to regulate NiMA independently of mitochondrial biogenesis. Inhibition of GSK3β with lithium or TWS119 stimulates NiMA in cultured human neurons, and mitochondrial activity and oxygen consumption in APP SAA mice. Conclusion NiMA disruption in vivo occurs before histopathological changes and cognitive decline in APP SAA mice, and may represent an early stage in human AD.
Collapse
|
11
|
Saberi R, Mirazi N, Amirahmadi S, Darbandi ZK, Vafaee F, Rajabian A, Hosseini M. Ameliorative effects of thiamin on learning behavior and memory dysfunction in a rat model of hypothyroidism: implication of oxidative stress and acetylcholinesterase. Metab Brain Dis 2023; 38:2603-2613. [PMID: 37906392 DOI: 10.1007/s11011-023-01317-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/23/2023] [Indexed: 11/02/2023]
Abstract
Hypothyroidism causes learning and memory impairment. Considering the neuroprotective properties of thiamine (Vitamin B1), this study was conducted to investigate the effects of thiamine on acetylcholinesterase (AChE) activity, oxidative damage, and memory deficits in hypothyroid rats.In this study, 50 rats (21 days old) were randomly divided into 5 groups and treated with propylthiouracil (0.05% in drinking water) and thiamine (50, 100, and 200 mg/kg, oral) for 7 weeks. Following that, Morris water maze (MWM) and passive avoidance (PA) tests were performed. Finally, oxidative stress indicators and AChE activity were measured in brain tissue.Treatment of hypothyroid rats with thiamine, especially at 100 and 200 mg/kg, alleviated the ability to remember the location of the platform as reflected by less time spent and distance to reach the platform, during the MWM test (P < 0.05 to P < 0.001). In the PA test, the latency to enter the dark chamber and light stay time were increased in rats who received thiamine compared to the hypothyroid group (P < 0.05 to P < 0.001). In addition, thiamine increased the levels of total thiol groups and superoxide dismutase while decreasing the levels of malondialdehyde and AChE.Our results suggest that thiamine supplementation could effectively improve memory loss in a rat model of hypothyroidism. The positive effects of thiamin on the learning and memory of hypothyroid rats may be due to amelioration of redox hemostasis and cholinergic disturbance.
Collapse
Affiliation(s)
- Rasul Saberi
- Department of Biology, Faculty of Basic Sciences, Bu-Ali Sina University, Hamedan, Iran
- Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Naser Mirazi
- Department of Biology, Faculty of Basic Sciences, Bu-Ali Sina University, Hamedan, Iran
| | - Sabiheh Amirahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Kioumarsi Darbandi
- Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzaneh Vafaee
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arezoo Rajabian
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mahmoud Hosseini
- Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
12
|
Murai T, Matsuda S. Metabolic Reprogramming toward Aerobic Glycolysis and the Gut Microbiota Involved in the Brain Amyloid Pathology. BIOLOGY 2023; 12:1081. [PMID: 37626967 PMCID: PMC10452252 DOI: 10.3390/biology12081081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023]
Abstract
Alzheimer's disease (AD) is characterized by the formation of senile plaques consisting of fibrillated amyloid-β (Aβ), dystrophic neurites, and the neurofibrillary tangles of tau. The oligomers/fibrillar Aβ damages the neurons or initiates an intracellular signaling cascade for neuronal cell death leading to Aβ toxicity. The Aβ is a 4 kDa molecular weight peptide originating from the C-terminal region of the amyloid precursor protein via proteolytic cleavage. Apart from the typical AD hallmarks, certain deficits in metabolic alterations have been identified. This study describes the emerging features of AD from the aspect of metabolic reprogramming in the main pathway of carbohydrate metabolism in the human brain. Particularly, the neurons in patients with AD favor glycolysis despite a normal mitochondrial function indicating a Warburg-like effect. In addition, certain dietary patterns are well known for their properties in preventing AD. Among those, a ketogenic diet may substantially improve the symptoms of AD. An effective therapeutic method for the treatment, mitigation, and prevention of AD has not yet been established. Therefore, the researchers pursue the development and establishment of novel therapies effective in suppressing AD symptoms and the elucidation of their underlying protective mechanisms against neurodegeneration aiming for AD therapy in the near future.
Collapse
Affiliation(s)
- Toshiyuki Murai
- Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita 565-0871, Japan;
| | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| |
Collapse
|
13
|
Boccardi V, Pigliautile M, Guazzarini AG, Mecocci P. The Potential of Fasting-Mimicking Diet as a Preventive and Curative Strategy for Alzheimer's Disease. Biomolecules 2023; 13:1133. [PMID: 37509169 PMCID: PMC10377404 DOI: 10.3390/biom13071133] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/05/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
This review examines the potential of fasting-mimicking diets (FMDs) in preventing and treating Alzheimer's disease (AD). FMDs are low-calorie diets that mimic the physiological and metabolic effects of fasting, including the activation of cellular stress response pathways and autophagy. Recent studies have shown that FMDs can reduce amyloid-beta accumulation, tau phosphorylation, and inflammation, as well as improve cognitive function in animal models of AD. Human studies have also reported improvements in AD biomarkers, cognitive functions, and subjective well-being measures following FMDs. However, the optimal duration and frequency of FMDs and their long-term safety and efficacy remain to be determined. Despite these uncertainties, FMDs hold promise as a non-pharmacological approach to AD prevention and treatment, and further research in this area is warranted.
Collapse
Affiliation(s)
- Virginia Boccardi
- Department of Medicine and Surgery, Institute of Gerontology and Geriatrics, University of Perugia, Piazzale Gambuli 1, 06132 Perugia, Italy
| | - Martina Pigliautile
- Department of Medicine and Surgery, Institute of Gerontology and Geriatrics, University of Perugia, Piazzale Gambuli 1, 06132 Perugia, Italy
| | - Anna Giulia Guazzarini
- Department of Medicine and Surgery, Institute of Gerontology and Geriatrics, University of Perugia, Piazzale Gambuli 1, 06132 Perugia, Italy
| | - Patrizia Mecocci
- Department of Medicine and Surgery, Institute of Gerontology and Geriatrics, University of Perugia, Piazzale Gambuli 1, 06132 Perugia, Italy
- Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, 17177 Stockholm, Sweden
| |
Collapse
|
14
|
Yang YH, Wen R, Yang N, Zhang TN, Liu CF. Roles of protein post-translational modifications in glucose and lipid metabolism: mechanisms and perspectives. Mol Med 2023; 29:93. [PMID: 37415097 DOI: 10.1186/s10020-023-00684-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/10/2023] [Indexed: 07/08/2023] Open
Abstract
The metabolism of glucose and lipids is essential for energy production in the body, and dysregulation of the metabolic pathways of these molecules is implicated in various acute and chronic diseases, such as type 2 diabetes, Alzheimer's disease, atherosclerosis (AS), obesity, tumor, and sepsis. Post-translational modifications (PTMs) of proteins, which involve the addition or removal of covalent functional groups, play a crucial role in regulating protein structure, localization function, and activity. Common PTMs include phosphorylation, acetylation, ubiquitination, methylation, and glycosylation. Emerging evidence indicates that PTMs are significant in modulating glucose and lipid metabolism by modifying key enzymes or proteins. In this review, we summarize the current understanding of the role and regulatory mechanisms of PTMs in glucose and lipid metabolism, with a focus on their involvement in disease progression associated with aberrant metabolism. Furthermore, we discuss the future prospects of PTMs, highlighting their potential for gaining deeper insights into glucose and lipid metabolism and related diseases.
Collapse
Affiliation(s)
- Yu-Hang Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, No.36, SanHao Street, Liaoning Province, Shenyang City, 110004, China
| | - Ri Wen
- Department of Pediatrics, Shengjing Hospital of China Medical University, No.36, SanHao Street, Liaoning Province, Shenyang City, 110004, China
| | - Ni Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, No.36, SanHao Street, Liaoning Province, Shenyang City, 110004, China
| | - Tie-Ning Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, No.36, SanHao Street, Liaoning Province, Shenyang City, 110004, China.
| | - Chun-Feng Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, No.36, SanHao Street, Liaoning Province, Shenyang City, 110004, China.
| |
Collapse
|
15
|
Thomas A, Ryan CP, Caspi A, Moffitt TE, Sugden K, Zhou J, Belsky DW, Gu Y. Diet, pace of biological aging, and risk of dementia in the Framingham Heart Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.24.23290474. [PMID: 37398353 PMCID: PMC10312831 DOI: 10.1101/2023.05.24.23290474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
INTRODUCTION We tested the hypothesis that healthy diet protects against dementia because it slows the pace of biological aging. METHODS We analyzed Framingham Offspring Cohort data (≥60y). We measured healthy diet using the Dietary Guideline for Americans (DGA, 3 visits 1991-2008), pace of aging using the DunedinPACE epigenetic clock (2005-2008), and incident dementia and mortality using records (compiled 2005-2018). RESULTS Of n=1,525 included participants (mean age 69.7, 54% female), n=129 developed dementia and n=432 died over follow-up. Greater DGA adherence was associated with slower DunedinPACE and reduced risks for dementia and mortality. Slower DunedinPACE was associated with reduced risks for dementia and mortality. Slower DunedinPACE accounted for 15% of the DGA association with dementia and 39% of the DGA association with mortality. DISCUSSION Findings suggest that slower pace of aging mediates part of the relationship of healthy diet with reduced dementia risk. Monitoring pace of aging may inform dementia prevention.
Collapse
Affiliation(s)
- Aline Thomas
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
| | - Calen P Ryan
- Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Avshalom Caspi
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Terrie E. Moffitt
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Karen Sugden
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Jiayi Zhou
- Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Daniel W. Belsky
- Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA
- Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Yian Gu
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
- Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University, New York, NY, USA
- Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA
| |
Collapse
|
16
|
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: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [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
|