1
|
Rajendra A, Bondonno NP, Murray K, Zhong L, Rainey-Smith SR, Gardener SL, Blekkenhorst LC, Doré V, Villemagne VL, Laws SM, Brown BM, Taddei K, Masters CL, Rowe CC, Martins RN, Hodgson JM, Bondonno CP. Baseline habitual dietary nitrate intake and Alzheimer's Disease related neuroimaging biomarkers in the Australian Imaging, Biomarkers and Lifestyle study of ageing. J Prev Alzheimers Dis 2025:100161. [PMID: 40221237 DOI: 10.1016/j.tjpad.2025.100161] [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: 10/23/2024] [Revised: 02/27/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Dietary nitrate, as a nitric oxide (NO) precursor, may support brain health and protect against dementia. OBJECTIVE Our primary aim was to investigate whether dietary nitrate is associated with neuroimaging markers of brain health linked with Alzheimer's disease (AD). PARTICIPANTS Study participants were cognitively unimpaired individuals from the Australian Imaging, Biomarkers and Lifestyle Study of Ageing (AIBL) who had β-amyloid positron emission tomography (PET) scans (n = 554) and magnetic resonance imaging (MRI) scans (n = 335) and had completed a Food Frequency Questionnaire at baseline. METHODS Source-specific nitrate intakes were estimated using comprehensive nitrate food composition databases. Rates of cerebral β-amyloid (Aβ) deposition, measured using PET, and rates of brain atrophy, measured using MRI, were assessed between baseline and 126-months follow-up, at intervals of 18 months. Multivariable-adjusted linear mixed effect models were used to examine associations between baseline source-specific nitrate intake and rates of (i) cerebral Aβ deposition and (ii) brain atrophy, over the 126 months of follow-up. Analyses were carried out following stratification of the sample by established dementia Alzheimer's disease (AD) risk factors including sex and presence or absence of the apolipoprotein E (APOE) ε4 allele. RESULTS In women carriers of the APOE ε4 allele, higher plant sourced nitrate intake (median intake 121 mg/day), was associated with a slower rate of cerebral Aβ deposition [β: 4.47 versus 8.99 Centiloid (CL) /18 months, p < 0.05] and right hippocampal atrophy [-0.01 versus -0.03 mm3 /18 months, p < 0.01], after multivariable adjustments. Moderate intake showed protective associations in men carriers and in both men and women non-carriers of APOE ε4. CONCLUSIONS Associations were observed between plant-derived nitrate intake and cerebral Aβ deposition, particularly in high-risk populations (women and APOE ε4 carriers). Associations were also observed for brain volume atrophy, however these exhibited subgroup variability without clear patterns relative to sex and APOE ε4 allele carriage. These findings suggest a potential link between plant-sourced nitrate and AD related neuroimaging markers of brain health improved brain health, but further validation in larger studies is required.
Collapse
Affiliation(s)
- Anjana Rajendra
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | - Nicola P Bondonno
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia; The Danish Cancer Institute, Copenhagen, Denmark
| | - Kevin Murray
- School of Population and Global Health, University of Western Australia, Perth, Western Australia, Australia
| | - Liezhou Zhong
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | - Stephanie R Rainey-Smith
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia; Lifestyle Approaches Towards Cognitive Health Research Group, Murdoch University, Murdoch, Western Australia, Australia; Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia; Australian Alzheimer's Research Foundation, Nedlands, Western Australia, Australia; School of Psychological Science, University of Western Australia, Perth, Western Australia, Australia
| | - Samantha L Gardener
- Lifestyle Approaches Towards Cognitive Health Research Group, Murdoch University, Murdoch, Western Australia, Australia; Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia; Australian Alzheimer's Research Foundation, Nedlands, Western Australia, Australia
| | - Lauren C Blekkenhorst
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia; For a full list of the AIBL Research Group see aibl.org.au
| | - Vincent Doré
- Australian E-Health Research Centre, CSIRO, 351 Royal Parade, Parkville, Victoria, Australia; Department of Molecular Imaging and Therapy, Austin Health, 145 Studley Road, Heidelberg, Victoria, Australia
| | - Victor L Villemagne
- Department of Molecular Imaging and Therapy, Austin Health, 145 Studley Road, Heidelberg, Victoria, Australia; Department of Psychiatry, University of Pittsburgh, Thomas Detre Hall, 3811 O'Hara Street, Pittsburgh, PA, USA; Centre for Precision Health, Edith Cowan University, 270 Joondalup Drive, Joondalup, Western Australia, Australia
| | - Simon M Laws
- Centre for Precision Health, Edith Cowan University, 270 Joondalup Drive, Joondalup, Western Australia, Australia; Collaborative Genomics and Translation Group, Edith Cowan University, 270 Joondalup Drive, Joondalup, Western Australia, Australia; Curtin Medical School, Curtin University, Kent Street, Bentley, Western Australia, Australia
| | - Belinda M Brown
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia; Lifestyle Approaches Towards Cognitive Health Research Group, Murdoch University, Murdoch, Western Australia, Australia; Collaborative Genomics and Translation Group, Edith Cowan University, 270 Joondalup Drive, Joondalup, Western Australia, Australia
| | - Kevin Taddei
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging and Therapy, Austin Health, 145 Studley Road, Heidelberg, Victoria, Australia; The Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Ralph N Martins
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Jonathan M Hodgson
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia; Medical School, The University of Western Australia, Royal Perth Hospital Research Foundation, Perth, Western Australia, Australia
| | - Catherine P Bondonno
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia; Medical School, The University of Western Australia, Royal Perth Hospital Research Foundation, Perth, Western Australia, Australia.
| |
Collapse
|
2
|
Wang X, Xin Z, Li X, Wu K, Wang W, Guo L, Wang L, Mo X, Liu X, Guo Z, Wang J, Lu C. Mediterranean diet and dementia: MRI marker evidence from meta-analysis. Eur J Med Res 2025; 30:32. [PMID: 39815306 PMCID: PMC11737277 DOI: 10.1186/s40001-025-02276-1] [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: 11/06/2024] [Accepted: 01/04/2025] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Dementia is a growing public health concern with limited effective treatments. Diet may be a modifiable factor that significantly impacts brain health. Mediterranean diet (MeDi) has been suggested to be associated with brain Magnetic Resonance Imaging (MRI) markers related to dementia, but the existing evidence is inconsistent. OBJECTIVES This systematic review and meta-analysis aimed to quantify the association between MeDi and dementia-related MRI markers. METHODS A systematic search was conducted on PubMed, Embase, and Web of Science up to September 2024. Two reviewers worked in parallel to select studies and extract data. We considered epidemiologic studies that reported beta coefficients (β) with 95% confidence intervals (CIs) for MRI markers related to MeDi. Separate meta-analyses were performed for cross-sectional and longitudinal studies. RESULTS A total of 20 relevant studies involving 44,893 individuals were included in the analysis. Thirteen cross-sectional studies included a total of 42,955 participants. A meta-analysis of cross-sectional studies revealed significant associations between MeDi and white matter hyperintensity (WMH) (β = - 0.03, 95% CI = - 0.05- - 0.01, P = 0.02). However, there were no significant associations found between MeDi and total brain volume (TBV) (β = - 0.03, 95% CI = - 0.20-0.13, P = 0.71), gray matter volume (GMV) (β = 0.26, 95% CI = - 0.19-0.71, P = 0.26), white matter volume (WMV) (β = - 0.09, 95% CI = - 0.40-0.22, P = 0.58), or hippocampal volume (HCV) (β = - 1.02, 95% CI = - 7.74-9.79, P = 0.82). In the longitudinal analysis, seven prospective studies with an average follow-up period ranging from 1.5 to 9 years and involving 1,938 participants. The combined effect size of MeDi showed no significant association with TBV or GMV. CONCLUSION Adherence to MeDi may be associated with reduced WMH in older adults. This suggests that MeDi may affect brain health and highlights the need for further research into its role as a modifiable lifestyle factor that might potentially modify the risk of dementia.
Collapse
Affiliation(s)
- Xiaojie Wang
- Department of Neurology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, 518067, Guangdong, China
| | - Zhiyao Xin
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen University, 74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, 510080, Guangdong, China
| | - Xiuwen Li
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen University, 74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, 510080, Guangdong, China
| | - Keying Wu
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen University, 74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, 510080, Guangdong, China
| | - Wanxin Wang
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen University, 74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, 510080, Guangdong, China
| | - Lan Guo
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen University, 74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, 510080, Guangdong, China
| | - Li Wang
- Department of Neurology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, 518067, Guangdong, China
| | - Xin Mo
- Department of Radiology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, 36 Industrial Road 7, Shenzhen, 518067, Guangdong, China
| | - Xinjian Liu
- Department of Neurology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, 518067, Guangdong, China
| | - Zhihui Guo
- Department of Neurology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, 518067, Guangdong, China
| | - Jing Wang
- Department of Radiology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, 36 Industrial Road 7, Shenzhen, 518067, Guangdong, China.
| | - Ciyong Lu
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-Sen University, 74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China.
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China.
- Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
3
|
Charisis S, Yannakoulia M, Scarmeas N. Diets to promote healthy brain ageing. Nat Rev Neurol 2025; 21:5-16. [PMID: 39572782 DOI: 10.1038/s41582-024-01036-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2024] [Indexed: 11/24/2024]
Abstract
Diet is a modifiable lifestyle factor with a proven role in cardiovascular disease risk reduction that might also play an important part in cognitive health. Evidence from observational studies has linked certain healthy dietary patterns to cognitive benefits. However, clinical trials of diet interventions have demonstrated either null or, at best, small effects on cognitive outcomes. In this Review, we summarize the currently available evidence from observational epidemiology and clinical trials regarding the potential role of diet in the prevention of cognitive decline and dementia. We further discuss possible methodological limitations that might have hindered the ability of previous diet intervention trials to capture potential neuroprotective effects. Considering the overwhelming and continuously expanding societal, economic and health-care burden of Alzheimer disease and other dementias, future nutritional research must address past methodological challenges to accurately and reliably inform clinical practice guidelines and public health policies. Within this scope, we provide a roadmap for future diet intervention trials for dementia prevention. We discuss study designs involving both intensive personalized interventions - to evaluate pharmacokinetic and pharmacodynamic properties, establish neuroprotective thresholds, and test hypothesized biological mechanisms and effects on brain health and cognition through sensitive and precise biomarker measures - and large-scale, pragmatic public health interventions to study population-level benefits.
Collapse
Affiliation(s)
- Sokratis Charisis
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
| | - Mary Yannakoulia
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | - Nikolaos Scarmeas
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece.
- The Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA.
- Taub Institute for Research in Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA.
| |
Collapse
|
4
|
Khandayataray P, Murthy MK. Dietary interventions in mitigating the impact of environmental pollutants on Alzheimer's disease - A review. Neuroscience 2024; 563:148-166. [PMID: 39542342 DOI: 10.1016/j.neuroscience.2024.11.020] [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: 08/25/2024] [Revised: 10/23/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Numerous studies linking environmental pollutants to oxidative stress, inflammation, and neurotoxicity have assigned pollutants to several neurodegenerative disorders, including Alzheimer's disease (AD). Heavy metals, pesticides, air pollutants, and endocrine disruptor chemicals have been shown to play important roles in AD development, with some traditional functions in amyloid-β formation, tau kinase action, and neuronal degeneration. However, pharmacological management and supplementation have resulted in limited improvement. This raises the interesting possibility that activities usually considered preventive, including diet, exercise, or mental activity, might be more similar to treatment or therapy for AD. This review focuses on the effects of diet on the effects of environmental pollutants on AD. One of the primary issues addressed in this review is a group of specific diets, including the Mediterranean diet (MeDi), Dietary Approaches to Stop Hypertension (DASH), and Mediterranean-DASH intervention for Neurodegenerative Delay (MIND), which prevent exposure to these toxins. Such diets have been proven to decrease oxidative stress and inflammation, which are unfavorable for neuronal growth. Furthermore, they contribute to positive changes in the composition of the human gut microbiota and thus encourage interactions in the Gut-Brain Axis, reducing inflammation caused by pollutants. This review emphasizes a multi-professional approach with reference to nutritional activities that would lower the neurotoxic load in populations with a high level of exposure to pollutants. Future studies focusing on diet and environment association plans may help identify preventive measures aimed at enhancing current disease deceleration.
Collapse
Affiliation(s)
- Pratima Khandayataray
- Department of Biotechnology, Academy of Management and Information Technology, Utkal University, Bhubaneswar, Odisha 752057, India
| | - Meesala Krishna Murthy
- Department of Allied Health Sciences, Chitkara School of Health Sciences, Chitkara University, Rajpura, Punjab 140401, India.
| |
Collapse
|
5
|
Ourry V, Binette AP, St-Onge F, Strikwerda-Brown C, Chagnot A, Poirier J, Breitner J, Arenaza-Urquijo EM, Rabin JS, Buckley R, Gonneaud J, Marchant NL, Villeneuve S. How Do Modifiable Risk Factors Affect Alzheimer's Disease Pathology or Mitigate Its Effect on Clinical Symptom Expression? Biol Psychiatry 2024; 95:1006-1019. [PMID: 37689129 DOI: 10.1016/j.biopsych.2023.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 08/11/2023] [Accepted: 09/03/2023] [Indexed: 09/11/2023]
Abstract
Epidemiological studies show that modifiable risk factors account for approximately 40% of the population variability in risk of developing dementia, including sporadic Alzheimer's disease (AD). Recent findings suggest that these factors may also modify disease trajectories of people with autosomal-dominant AD. With positron emission tomography imaging, it is now possible to study the disease many years before its clinical onset. Such studies can provide key knowledge regarding pathways for either the prevention of pathology or the postponement of its clinical expression. The former "resistance pathway" suggests that modifiable risk factors could affect amyloid and tau burden decades before the appearance of cognitive impairment. Alternatively, the resilience pathway suggests that modifiable risk factors may mitigate the symptomatic expression of AD pathology on cognition. These pathways are not mutually exclusive and may appear at different disease stages. Here, in a narrative review, we present neuroimaging evidence that supports both pathways in sporadic AD and autosomal-dominant AD. We then propose mechanisms for their protective effect. Among possible mechanisms, we examine neural and vascular mechanisms for the resistance pathway. We also describe brain maintenance and functional compensation as bases for the resilience pathway. Improved mechanistic understanding of both pathways may suggest new interventions.
Collapse
Affiliation(s)
- Valentin Ourry
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada.
| | - Alexa Pichet Binette
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada; Clinical Memory Research Unit, Department of Clinical Sciences, Lunds Universitet, Malmö, Sweden
| | - Frédéric St-Onge
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Cherie Strikwerda-Brown
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada; School of Psychological Science, The University of Western Australia, Perth, Western Australia, Australia
| | - Audrey Chagnot
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Judes Poirier
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - John Breitner
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Eider M Arenaza-Urquijo
- Environment and Health over the Lifecourse Programme, Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain; Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Jennifer S Rabin
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada; Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada; Rehabilitation Sciences Institute, University of Toronto, Toronto, Ontario, Canada
| | - Rachel Buckley
- Melbourne School of Psychological Sciences University of Melbourne, Parkville, Victoria, Australia; Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Julie Gonneaud
- Normandie University, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Natalie L Marchant
- Division of Psychiatry, University College London, London, United Kingdom
| | - Sylvia Villeneuve
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada; McConnell Brain Imaging Center, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
6
|
Grasset L, Planche V, Bouteloup V, Azouani C, Dubois B, Blanc F, Paquet C, David R, Belin C, Jonveaux T, Julian A, Pariente J, Mangin JF, Chêne G, Dufouil C. Physical activity, biomarkers of brain pathologies and dementia risk: Results from the Memento clinical cohort. Alzheimers Dement 2023; 19:5700-5718. [PMID: 37422285 DOI: 10.1002/alz.13360] [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: 01/20/2023] [Revised: 05/25/2023] [Accepted: 05/29/2023] [Indexed: 07/10/2023]
Abstract
INTRODUCTION This study aims to examine whether physical activity moderates the association between biomarkers of brain pathologies and dementia risk. METHODS From the Memento cohort, we analyzed 1044 patients with mild cognitive impairment, aged 60 and older. Self-reported physical activity was assessed using the International Physical Activity Questionnaire. Biomarkers of brain pathologies comprised medial temporal lobe atrophy (MTA), white matter lesions, and plasma amyloid beta (Aβ)42/40 and phosphorylated tau181. Association between physical activity and risk of developing dementia over 5 years of follow-up, and interactions with biomarkers of brain pathologies were tested. RESULTS Physical activity moderated the association between MTA and plasma Aβ42/40 level and increased dementia risk. Compared to participants with low physical activity, associations of both MTA and plasma Aβ42/40 on dementia risk were attenuated in participants with high physical activity. DISCUSSION Although reverse causality cannot be excluded, this work suggests that physical activity may contribute to cognitive reserve. HIGHLIGHTS Physical activity is an interesting modifiable target for dementia prevention. Physical activity may moderate the impact of brain pathology on dementia risk. Medial temporal lobe atrophy and plasma amyloid beta 42/40 ratio were associated with increased dementia risk especially in those with low level of physical activity.
Collapse
Affiliation(s)
- Leslie Grasset
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219, CIC1401-EC, Bordeaux, France
| | - Vincent Planche
- University of Bordeaux, CNRS UMR 5293, Institut des Maladies Neurodégénératives, Centre Mémoire de Ressources et de Recherches, Pôle de Neurosciences Cliniques, CHU de Bordeaux, Bordeaux, France
| | - Vincent Bouteloup
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219, CIC1401-EC, Bordeaux, France
- Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux, Bordeaux, France
| | - Chabha Azouani
- CATI multicentre imaging platform, US52-UAR2031, CEA, ICM, SU, CNRS, INSERM, APHP, Gif-sur-Yvette, France
| | - Bruno Dubois
- IM2A AP-HP INSERM UMR-S975 Groupe Hospitalier Pitié-Salpêtrière Institut de la Mémoire et de la Maladie d'Alzheimer Institut du Cerveau et de la Moelle épinière Sorbonne Université Paris, Paris, France
| | - Frédéric Blanc
- ICube laboratory, Pôle de Gériatrie, Université de Strasbourg, CNRS, UMR 7357, Fédération de Médecine Translationnelle de Strasbourg, Centre Mémoire de Ressources et de Recherches, Strasbourg, France
| | - Claire Paquet
- Université de Paris Cité, Centre de Neurologie Cognitive GHU APHP Nord Hôpital Lariboisière, INSERMU1144, Paris, France
| | - Renaud David
- Department of Old Age Psychiatry, Nice University Hospital, Nice, France
| | - Catherine Belin
- Service de Neurologie Hôpital Saint-Louis AP-HP, Paris, France
| | - Thérèse Jonveaux
- Centre Mémoire de Ressources et de Recherche de Lorraine, Service de Neurologie CHRU Nancy, Laboratoire Lorrain de Psychologie et de Neurosciences de la dynamique des comportements 2LPN EA 7489 Université de Lorraine, Nancy, France
| | - Adrien Julian
- Service de Neurologie CHU La Milétrie Centre Mémoire de Ressources et de Recherche, Poitiers, France
- Centre d'Investigation Clinique CIC1402, Poitiers, France
| | - Jérémie Pariente
- Department of Neurology, Toulouse University Hospital, Toulouse, France
- Toulouse NeuroImaging Center, Universite de Toulouse, Inserm, UPS, Toulouse, France
| | - Jean-François Mangin
- CATI multicentre imaging platform, US52-UAR2031, CEA, ICM, SU, CNRS, INSERM, APHP, Gif-sur-Yvette, France
- Université Paris-Saclay, CEA, CNRS, Neurospin, UMR 9027, Gif-sur-Yvette, France
| | - Geneviève Chêne
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219, CIC1401-EC, Bordeaux, France
- Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux, Bordeaux, France
| | - Carole Dufouil
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219, CIC1401-EC, Bordeaux, France
- Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux, Bordeaux, France
| |
Collapse
|
7
|
Al-darsani Z, Jacobs DR, Bryan RN, Launer LJ, Steffen LM, Yaffe K, Shikany JM, Odegaard AO. Measures of MRI Brain Biomarkers in Middle Age According to Average Modified Mediterranean Diet Scores Throughout Young and Middle Adulthood. NUTRITION AND HEALTHY AGING 2023; 8:109-121. [PMID: 38013773 PMCID: PMC10475985 DOI: 10.3233/nha-220192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 06/08/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND The Mediterranean diet (MedDiet) has been linked with better cognitive function and brain integrity. OBJECTIVE To examine the association of modified Mediterranean diet (mMedDiet) scores from early through middle adulthood in relation to volumetric and microstructural midlife MRI brain measures. Assess the association of mMedDiet and brain measures with four cognitive domains. If variables are correlated, determine if brain measures mediate the relationship between mMedDiet and cognition. METHODS 618 participants (mean age 25.4±3.5 at year 0) of the Coronary Artery Risk Development in Young Adults (CARDIA) study were included. Cumulative average mMedDiet scores were calculated by averaging scores from years 0, 7, and 20. MRI scans were obtained at years 25 and 30. General linear models were used to examine the association between mMedDiet and brain measures. RESULTS Higher cumulative average mMedDiet scores were associated with better microstructural white matter (WM) integrity measured by fractional anisotropy (FA) at years 25 and 30 (all ptrend <0.05). Higher mMedDiet scores at year 7 were associated with higher WM FA at year 25 (β= 0.003, ptrend = 0.03). Higher mMedDiet scores at year 20 associated with higher WM FA at years 25 (β= 0.0005, ptrend = 0.002) and 30 (β= 0.0003, ptrend = 0.02). mMedDiet scores were not associated with brain volumes. Higher mMedDiet scores and WM FA were both correlated with better executive function, processing speed, and global cognition (all ptrend <0.05). WM FA did not mediate the association between mMedDiet scores and cognition. CONCLUSIONS mMedDiet scores may be associated with microstructural WM integrity at midlife.
Collapse
Affiliation(s)
- Zeinah Al-darsani
- Department of Epidemiology and Biostatistics, University of California, Irvine, Irvine, CA, USA
| | - David R. Jacobs
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - R. Nick Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Lenore J. Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Baltimore, MD, USA
| | - Lyn M. Steffen
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Kristine Yaffe
- Department of Psychiatry, Neurology, and Epidemiology and Biostatistics, University of California, San Francisco, USA
| | - James M. Shikany
- Division of Preventive Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Andrew O. Odegaard
- Department of Epidemiology and Biostatistics, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
8
|
Agarwal P, Leurgans SE, Agrawal S, Aggarwal NT, Cherian LJ, James BD, Dhana K, Barnes LL, Bennett DA, Schneider JA. Association of Mediterranean-DASH Intervention for Neurodegenerative Delay and Mediterranean Diets With Alzheimer Disease Pathology. Neurology 2023; 100:e2259-e2268. [PMID: 36889921 PMCID: PMC10259273 DOI: 10.1212/wnl.0000000000207176] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 01/26/2023] [Indexed: 03/10/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Diet may reduce Alzheimer dementia risk and slow cognitive decline, but the understanding of the relevant neuropathologic mechanisms remains limited. The association of dietary patterns with Alzheimer disease (AD) pathology has been suggested using neuroimaging biomarkers. This study examined the association of Mediterranean-DASH Intervention for Neurodegenerative Delay (MIND) and Mediterranean dietary patterns with β-amyloid load, phosphorylated tau tangles, and global AD pathology in postmortem brain tissue of older adults. METHODS Autopsied participants of the Rush Memory and Aging Project with complete dietary information (collected through a validated food frequency questionnaire) and AD pathology data (β-amyloid load, phosphorylated tau tangles, and global AD pathology [summarized neurofibrillary tangles and neuritic and diffuse plaques]) were included in this study. Linear regression models controlled for age at death, sex, education, APOE-ε4 status, and total calories were used to investigate the dietary patterns (MIND and Mediterranean diets) and dietary components associated with AD pathology. Further effect modification was tested for APOE-ε4 status and sex. RESULTS Among our study participants (N = 581, age at death: 91.0 ± 6.3 years; mean age at first dietary assessment: 84.2 ± 5.8 years; 73% female; 6.8 ± 3.9 years of follow-up), dietary patterns were associated with lower global AD pathology (MIND: β = -0.022, p = 0.034, standardized β = -2.0; Mediterranean: β = -0.007, p = 0.039, standardized β = -2.3) and specifically less β-amyloid load (MIND: β = -0.068, p = 0.050, standardized β = -2.0; Mediterranean: β = -0.040, p = 0.004, standardized β = -2.9). The findings persisted when further adjusted for physical activity, smoking, and vascular disease burden. The associations were also retained when participants with mild cognitive impairment or dementia at the baseline dietary assessment were excluded. Those in the highest tertile of green leafy vegetables intake had less global AD pathology when compared with those in the lowest tertile (tertile 3 vs tertile 1: β = -0.115, p = 0.0038). DISCUSSION The MIND and Mediterranean diets are associated with less postmortem AD pathology, primarily β-amyloid load. Among dietary components, higher green leafy vegetable intake was associated with less AD pathology.
Collapse
Affiliation(s)
- Puja Agarwal
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL.
| | - Sue E Leurgans
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL
| | - Sonal Agrawal
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL
| | - Neelum T Aggarwal
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL
| | - Laurel J Cherian
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL
| | - Bryan D James
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL
| | - Klodian Dhana
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL
| | - Lisa L Barnes
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL
| | - David A Bennett
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL
| | - Julie A Schneider
- From the Rush Alzheimer's Disease Center (P.A., S.E.L., N.A., B.D.J., L.L.B., D.A.B., J.A.S.), Departments of Internal Medicine (P.A., B.D.J., K.D.), Clinical Nutrition (P.A.), Neurological Sciences (S.E.L., N.T.A., L.J.C., L.L.B., D.A.B., J.A.S.) and Pathology (S.A., J.A.S.); and Rush Institute of Healthy Aging (K.D.), Rush University Medical Center, Chicago, IL
| |
Collapse
|
9
|
Hwang J, Choe YM, Suh GH, Lee BC, Choi IG, Lee JH, Kim HS, Kim SG, Yi D, Kim JW. Spicy food intake predicts Alzheimer-related cognitive decline in older adults with low physical activity. Sci Rep 2023; 13:7942. [PMID: 37193785 PMCID: PMC10188510 DOI: 10.1038/s41598-023-35234-0] [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: 02/01/2023] [Accepted: 05/15/2023] [Indexed: 05/18/2023] Open
Abstract
A plausible association exists among spicy food consumption, physical activity, and Alzheimer's disease (AD) or cognitive decline, but it remains poorly investigated. We aimed to examined the association between spicy food and AD-related memory decline or global cognitive decline in older adults under the moderating effect of physical activity. Total 196 non-demented older adults were included. Participants underwent comprehensive dietary and clinical assessments including spicy food intake, AD-related memory, global cognition, and physical activity. The strength of spicy food was stratified into three categories: 'not spicy' (reference), 'low spiciness', and 'high spiciness'. Multiple linear regression analyses were performed to examine the relationships between spicy level and cognition. The spicy level was the independent variable in each analysis; it was entered as a stratified categorical variable using the three categories. We found a significant association between a high level of spiciness in food and decreased memory ([Formula: see text] - 0.167, p < 0.001) or global cognition ([Formula: see text] - 0.122, p = 0.027), but not non-memory cognition. To explore the moderating effects of age, sex, apolipoprotein E ε4 allele-positivity, vascular risk score, body mass index, and physical activity on the associations between spicy level and memory or global cognition, the same regression analyses were repeated including two-way interaction terms between the spicy level and each of the six variables as an additional independent variable. An interactive effect was detected between a high level of spiciness in food and physical activity on the memory ([Formula: see text] 0.209, p = 0.029) or global cognition ([Formula: see text] 0.336, p = 0.001). Subgroup analyses showed that the association between a high level of spiciness in food and a lower memory ([Formula: see text] - 0.254, p < 0.001) and global score ([Formula: see text] - 0.222, p = 0.002) was present only in older adults with low physical activity, but not in older adults with high physical activity. Our findings suggest that spicy food intake is predictive of AD-related cognitive decline, i.e., episodic memory; this relationship is worsened by physically inactive lifestyle.
Collapse
Affiliation(s)
- Jaeuk Hwang
- Department of Psychiatry, Soonchunhyang University Hospital Seoul, Seoul, 04401, Republic of Korea
| | - Young Min Choe
- Department of Neuropsychiatry, Hallym University Dongtan Sacred Heart Hospital, 7 Keunjaebong-gil, Hwaseong-si, Gyeonggi-do, 18450, Republic of Korea
- Department of Psychiatry, Hallym University College of Medicine, Chuncheon, Gangwon, 24252, Republic of Korea
| | - Guk-Hee Suh
- Department of Neuropsychiatry, Hallym University Dongtan Sacred Heart Hospital, 7 Keunjaebong-gil, Hwaseong-si, Gyeonggi-do, 18450, Republic of Korea
- Department of Psychiatry, Hallym University College of Medicine, Chuncheon, Gangwon, 24252, Republic of Korea
| | - Boung Chul Lee
- Department of Psychiatry, Hallym University College of Medicine, Chuncheon, Gangwon, 24252, Republic of Korea
- Department of Neuropsychiatry, Hallym University Hangang Sacred Heart Hospital, Seoul, 07247, Republic of Korea
| | - Ihn-Geun Choi
- Department of Psychiatry, Hallym University College of Medicine, Chuncheon, Gangwon, 24252, Republic of Korea
- Department of Psychiatry, Seoul W Psychiatric Office, Seoul, 07247, Republic of Korea
| | - Jun Ho Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, 03080, Republic of Korea
| | - Hyun Soo Kim
- Department of Laboratory Medicine, Hallym University Dongtan Sacred Heart Hospital, 7 Keunjaebong-gil, Hwaseong-si, Gyeonggi-do, 18450, Republic of Korea
| | - Shin Gyeom Kim
- Department of Neuropsychiatry, Soonchunhyang University Bucheon Hospital, Bucheon, 14584, Republic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, 03080, Republic of Korea
| | - Jee Wook Kim
- Department of Neuropsychiatry, Hallym University Dongtan Sacred Heart Hospital, 7 Keunjaebong-gil, Hwaseong-si, Gyeonggi-do, 18450, Republic of Korea.
- Department of Psychiatry, Hallym University College of Medicine, Chuncheon, Gangwon, 24252, Republic of Korea.
| |
Collapse
|
10
|
Arnoldy L, Gauci S, Young LM, Marx W, Macpherson H, Pipingas A, Civier O, White DJ. The association of dietary and nutrient patterns on neurocognitive decline: A systematic review of MRI and PET studies. Ageing Res Rev 2023; 87:101892. [PMID: 36878405 DOI: 10.1016/j.arr.2023.101892] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 02/14/2023] [Accepted: 02/27/2023] [Indexed: 03/08/2023]
Abstract
BACKGROUND As the global population ages, there has been a growing incidence of neurodegenerative diseases such as Alzheimer's. More recently, studies exploring the relationship between dietary patterns and neuroimaging outcomes have received particular attention. This systematic literature review provides a structured overview of the association between dietary and nutrient patterns on neuroimaging outcomes and cognitive markers in middle-aged to older adults. A comprehensive literature search was conducted to find relevant articles published from 1999 to date using the following databases Ovid MEDLINE, Embase, PubMed, Scopus and Web of Science. The inclusion criteria for the articles comprised studies reporting on the association between dietary patterns and neuroimaging outcomes, which includes both specific pathological hallmarks of neurodegenerative diseases such as Aβ and tau and nonspecific markers such as structural MRI and glucose metabolism. The risk of bias was evaluated using the Quality Assessment tool from the National Heart, Lung, and Blood Institute of the National Institutes of Health. The results were then organized into a summary of results table, collated based on synthesis without meta-analysis. After conducting the search, 6050 records were extracted and screened for eligibility, with 107 eligible for full-text screening and 42 articles ultimately being included in this review. The results of the systematic review indicate that there is some evidence suggesting that healthy dietary and nutrient patterns were associated with neuroimaging measures, indicative of a protective influence on neurodegeneration and brain ageing. Conversely, unhealthy dietary and nutrient patterns showed evidence pointing to decreased brain volumes, poorer cognition and increased Aβ deposition. Future research should focus on sensitive neuroimaging acquisition and analysis methods, to study early neurodegenerative changes and identify critical periods for interventions and prevention. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no, CRD42020194444).
Collapse
Affiliation(s)
- Lizanne Arnoldy
- Centre of Human Psychopharmacology, Centre for Mental Health and Brain Sciences, Swinburne University, Melbourne Australia.
| | - Sarah Gauci
- Centre of Human Psychopharmacology, Centre for Mental Health and Brain Sciences, Swinburne University, Melbourne Australia; IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Deakin University, Geelong, Australia
| | - Lauren M Young
- Centre of Human Psychopharmacology, Centre for Mental Health and Brain Sciences, Swinburne University, Melbourne Australia; IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Deakin University, Geelong, Australia
| | - Wolfgang Marx
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Deakin University, Geelong, Australia
| | - Helen Macpherson
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, Australia
| | - Andrew Pipingas
- Centre of Human Psychopharmacology, Centre for Mental Health and Brain Sciences, Swinburne University, Melbourne Australia
| | - Oren Civier
- Swinburne Neuroimaging, Swinburne University, Melbourne, Australia
| | - David J White
- Centre of Human Psychopharmacology, Centre for Mental Health and Brain Sciences, Swinburne University, Melbourne Australia; Swinburne Neuroimaging, Swinburne University, Melbourne, Australia
| |
Collapse
|
11
|
Yassine HN, Self W, Kerman BE, Santoni G, Navalpur Shanmugam N, Abdullah L, Golden LR, Fonteh AN, Harrington MG, Gräff J, Gibson GE, Kalaria R, Luchsinger JA, Feldman HH, Swerdlow RH, Johnson LA, Albensi BC, Zlokovic BV, Tanzi R, Cunnane S, Samieri C, Scarmeas N, Bowman GL. Nutritional metabolism and cerebral bioenergetics in Alzheimer's disease and related dementias. Alzheimers Dement 2023; 19:1041-1066. [PMID: 36479795 PMCID: PMC10576546 DOI: 10.1002/alz.12845] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/12/2022] [Accepted: 10/05/2022] [Indexed: 12/13/2022]
Abstract
Disturbances in the brain's capacity to meet its energy demand increase the risk of synaptic loss, neurodegeneration, and cognitive decline. Nutritional and metabolic interventions that target metabolic pathways combined with diagnostics to identify deficits in cerebral bioenergetics may therefore offer novel therapeutic potential for Alzheimer's disease (AD) prevention and management. Many diet-derived natural bioactive components can govern cellular energy metabolism but their effects on brain aging are not clear. This review examines how nutritional metabolism can regulate brain bioenergetics and mitigate AD risk. We focus on leading mechanisms of cerebral bioenergetic breakdown in the aging brain at the cellular level, as well as the putative causes and consequences of disturbed bioenergetics, particularly at the blood-brain barrier with implications for nutrient brain delivery and nutritional interventions. Novel therapeutic nutrition approaches including diet patterns are provided, integrating studies of the gut microbiome, neuroimaging, and other biomarkers to guide future personalized nutritional interventions.
Collapse
Affiliation(s)
- Hussein N Yassine
- Department of Medicine, Keck School of Medicine, University of Southern, California, Los Angeles, California, USA
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Wade Self
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Bilal E Kerman
- Department of Medicine, Keck School of Medicine, University of Southern, California, Los Angeles, California, USA
| | - Giulia Santoni
- Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale Lausanne (EPFL), Lausanne, Switzerland
| | - NandaKumar Navalpur Shanmugam
- Department of Neurology, Genetics and Aging Research Unit, McCance Center for Brain Health, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Lesley R Golden
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Alfred N Fonteh
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Huntington Medical Research Institutes, Pasadena, California, USA
| | - Michael G Harrington
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Johannes Gräff
- Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale Lausanne (EPFL), Lausanne, Switzerland
| | - Gary E Gibson
- Brain and Mind Research Institute, Weill Cornell Medicine, Burke Neurological Institute, White Plains, New York, USA
| | - Raj Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Jose A Luchsinger
- Department of Medicine and Epidemiology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Howard H Feldman
- Department of Neurosciences, University of California, San Diego, California, USA
| | - Russell H Swerdlow
- Department of Neurology, University of Kansas School of Medicine, Kansas City, Kansas, USA
| | - Lance A Johnson
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Benedict C Albensi
- Nova Southeastern Univ. College of Pharmacy, Davie, Florida, USA
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Rudolph Tanzi
- Department of Neurology, Genetics and Aging Research Unit, McCance Center for Brain Health, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Stephen Cunnane
- Department of Medicine, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Cécilia Samieri
- Univ. Bordeaux, INSERM, BPH, U1219, F-33000, Bordeaux, France
| | - Nikolaos Scarmeas
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
- Department of Neurology, Columbia University, New York City, New York, USA
| | - Gene L Bowman
- Department of Neurology, Genetics and Aging Research Unit, McCance Center for Brain Health, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Helfgott Research Institute, National University of Natural Medicine, Portland, Oregon, USA
| |
Collapse
|
12
|
Devranis P, Vassilopoulou Ε, Tsironis V, Sotiriadis PM, Chourdakis M, Aivaliotis M, Tsolaki M. Mediterranean Diet, Ketogenic Diet or MIND Diet for Aging Populations with Cognitive Decline: A Systematic Review. LIFE (BASEL, SWITZERLAND) 2023; 13:life13010173. [PMID: 36676122 PMCID: PMC9866105 DOI: 10.3390/life13010173] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 01/10/2023]
Abstract
(1) Background: Compelling evidence shows that dietary patterns can slow the rate of cognitive decline, suggesting diet is a promising preventive measure against dementia. (2) Objective: This systematic review summarizes the evidence of three dietary patterns, the Mediterranean diet, the ketogenic diet and the MIND diet, for the prevention of cognitive decline. (3) Methods: A systematic search was conducted in major electronic databases (PubMed, ScienceDirect and Web of Science) up until 31 January 2022, using the key search terms "Mediterranean diet", "ketogenic diet", "MIND diet", "dementia", "cognition" and "aging". A statistical analysis was performed using RoB 2 and the Jadad scale to assess the risk of bias and methodological quality in randomized controlled trials. (4) Results: Only RCTs were included in this study; there were eleven studies (n = 2609 participants) of the Mediterranean diet, seven studies (n = 313) of the ketogenic diet and one study (n = 37) of the MIND diet. The participants' cognitive statuses were normal in seven studies, ten studies included patients with mild cognitive impairments and two studies included Alzheimer's disease patients. (5) Conclusion: All three dietary interventions have been shown to slow the rate of cognitive decline in the included studies. The Mediterranean diet was shown to be beneficial for global cognition after 10 weeks of adherence, the ketogenic diet had a beneficial effect for patients with diabetes mellitus and improved verbal recognition, while the MIND diet showed benefits in obese patients, improving working memory, verbal recognition, memory and attention.
Collapse
Affiliation(s)
- Paschalis Devranis
- 1st Department of Neurology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA University Hospital, 54636 Thessaloniki, Greece
| | - Εmilia Vassilopoulou
- Department of Nutritional Sciences and Dietetics, International Hellenic University, 57400 Thessaloniki, Greece
| | - Vasileios Tsironis
- Department of Nutritional Sciences and Dietetics, International Hellenic University, 57400 Thessaloniki, Greece
| | | | - Michail Chourdakis
- Laboratory of Hygiene, Social & Preventive Medicine and Medical Statistics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Michalis Aivaliotis
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Magdalini Tsolaki
- 1st Department of Neurology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA University Hospital, 54636 Thessaloniki, Greece
- Greek Alzheimer Association and Related Disorders, 54643 Thessaloniki, Greece
| |
Collapse
|
13
|
Díaz G, Lengele L, Sourdet S, Soriano G, de Souto Barreto P. Nutrients and amyloid β status in the brain: A narrative review. Ageing Res Rev 2022; 81:101728. [PMID: 36049590 DOI: 10.1016/j.arr.2022.101728] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/18/2022] [Accepted: 08/26/2022] [Indexed: 01/31/2023]
Abstract
Amyloid beta (Aβ) is a peptide and a hallmark of Alzheimer's disease (AD). Emerging evidence suggests that Aβ levels could be influenced by diet. However, the evidence is sparse and for some nutrients, controversial. The aim of this narrative review is to gather the findings of observational and clinical trials involving human participants on the relationships between nutrients and brain Aβ status. Some dietary patterns are associated to reduced levels of Aβ in the brain, such as the Mediterranean diet, ketogenic diet as well as low intake of saturated fat, high-glycemic-index food, sodium, and junk/fast food. Low Aβ status in the brain was also associated with higher density lipoproteins (HDL) cholesterol and polyunsaturated fatty acids consumption. Data on alcohol intake is not conclusive. On the contrary, high Aβ levels in the brain were related to a higher intake of total cholesterol, triglycerides, low-density lipoproteins (LDL) cholesterol, saturated fat, sucrose, and fructose. Folic acid, cobalamin, vitamin E, and vitamin D were not associated to Aβ status, while high blood concentrations of Calcium, Aluminum, Zinc, Copper, and Manganese were associated with decreased Aβ blood levels but were not associated with Aβ cerebral spinal fluid (CSF) concentrations. In conclusion, certain dietary patterns and nutrients are associated to brain Aβ status. Further research on the association between nutrients and brain Aβ status is needed in order to pave the way to use nutritional interventions as efficacious strategies to prevent Aβ disturbance and potentially AD.
Collapse
Affiliation(s)
- Gustavo Díaz
- Faculty of Medicine, Research Institute on Nutrition, Genetics, and Metabolism, Universidad El Bosque, Bogotá, Colombia; Research In Colombia Foundation, Bogotá, Colombia.
| | - Laetitia Lengele
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo Universitaire de Toulouse, France
| | - Sandrine Sourdet
- Gérontopôle, Department of Internal Medicine and Geriatrics, Toulouse University Hospital, La Cité de la Santé, Hôpital La Grave, Place Lange, Cedex 9, TSA 60033, Toulouse 31059, France
| | - Gaëlle Soriano
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo Universitaire de Toulouse, France; Gérontopôle, Department of Internal Medicine and Geriatrics, Toulouse University Hospital, La Cité de la Santé, Hôpital La Grave, Place Lange, Cedex 9, TSA 60033, Toulouse 31059, France
| | - Philipe de Souto Barreto
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo Universitaire de Toulouse, France; UPS/Inserm, CERPOP UMR1295, University of Toulouse III, Toulouse, France
| |
Collapse
|
14
|
Felisatti F, Gonneaud J, Palix C, Garnier-Crussard A, Mézenge F, Landeau B, Chocat A, Quillard A, Ferrand-Devouge E, de La Sayette V, Vivien D, Chételat G, Poisnel G. Role of Cardiovascular Risk Factors on the Association Between Physical Activity and Brain Integrity Markers in Older Adults. Neurology 2022; 98:e2023-e2035. [PMID: 35418459 PMCID: PMC9162049 DOI: 10.1212/wnl.0000000000200270] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 02/08/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Physical activity has been associated with a decreased risk for dementia, but the mechanisms underlying this association remain to be determined. Our objective was to assess whether cardiovascular risk factors mediate the association between physical activity and brain integrity markers in older adults. METHODS At baseline, participants from the Age-Well study completed a physical activity questionnaire and underwent cardiovascular risk factors collection (systolic blood pressure, body mass index [BMI], current smoker status, and high-density lipoprotein cholesterol, total cholesterol, and insulin levels) and multimodal neuroimaging (structural MRI, diffusion MRI, FDG-PET, and florbetapir PET). Multiple regressions were conducted to assess the association among physical activity, cardiovascular risk factors, and neuroimaging. Mediation analyses were performed to test whether cardiovascular risk factors mediated the associations between physical activity and neuroimaging. RESULTS A total of 134 cognitively unimpaired older adults (≥65 years) were included. Higher physical activity was associated with higher gray matter (GM) volume (β = 0.174, p = 0.030) and cerebral glucose metabolism (β = 0.247, p = 0.019) but not with amyloid deposition or white matter integrity. Higher physical activity was associated with lower insulin level and BMI but not with the other cardiovascular risk factors. Lower insulin level and BMI were related to higher GM volume but not to cerebral glucose metabolism. When controlling for insulin level and BMI, the association between physical activity and cerebral glucose metabolism remained unchanged, while the association with GM volume was lost. When insulin level and BMI were entered in the same model, only BMI remained a significant predictor of GM volume. Mediation analyses confirmed that insulin level and BMI mediated the association between physical activity and GM volume. Analyses were replicated within Alzheimer disease-sensitive regions and results remained overall similar. DISCUSSION The association between physical activity and GM volume is mediated by changes in insulin level and BMI. In contrast, the association with cerebral glucose metabolism seems to be independent from cardiovascular risk factors. Older adults engaging in physical activity experience cardiovascular benefits through the maintenance of a lower BMI and insulin level, resulting in greater structural brain integrity. This study has implications for understanding how physical activity affects brain health and may help in developing strategies to prevent or delay age-related decline. TRIAL REGISTRATION INFORMATION EudraCT: 2016-002,441-36; IDRCB: 2016-A01767-44; ClinicalTrials.gov Identifier: NCT02977819.
Collapse
Affiliation(s)
- Francesca Felisatti
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Julie Gonneaud
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Cassandre Palix
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Antoine Garnier-Crussard
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Florence Mézenge
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Brigitte Landeau
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Anne Chocat
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Anne Quillard
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Eglantine Ferrand-Devouge
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Vincent de La Sayette
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Denis Vivien
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Gaël Chételat
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| | - Géraldine Poisnel
- From PhIND, "Physiopathology and Imaging of Neurological Disorders" (F.F., J.G., C.P., A.G.-C., F.M., B.L., A.C., A.Q., E.F.-D., D.V., G.C., G.P.), Institut Blood and Brain at Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen; Clinical and Research Memory Center of Lyon (A.G.-C.), Lyon Institute For Elderly, Charpennes Hospital, Hospices Civils de Lyon; Claude Bernard University Lyon 1 (A.G.-C.); Department of General Practice (E.F.-D.), Normandie Univ, UNIROUEN, Rouen; Rouen University Hospital, CIC-CRB 1404 (E.F.-D.); PSL Université, EPHE (V.d.L.S.), Normandie Univ, UNICAEN, INSERM, U1077, CHU de Caen, GIP Cyceron, NIMH; and Département de Recherche Clinique (D.V.), CHU Caen-Normandie, Caen, France
| |
Collapse
|
15
|
Townsend RF, Woodside JV, Prinelli F, O'Neill RF, McEvoy CT. Associations Between Dietary Patterns and Neuroimaging Markers: A Systematic Review. Front Nutr 2022; 9:806006. [PMID: 35571887 PMCID: PMC9097077 DOI: 10.3389/fnut.2022.806006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 03/16/2022] [Indexed: 12/13/2022] Open
Abstract
Dementia is a complex, growing challenge for population health worldwide. Dietary patterns (DPs) may offer an opportunity to beneficially influence cognitive ageing and potentially reduce an individuals’ risk of dementia through diet-related mechanisms. However, previous studies within this area have shown mixed results, which may be partly explained by the lack of sensitivity and accuracy within cognitive testing methods. Novel neuroimaging techniques provide a sensitive method to analyse brain changes preceding cognitive impairment which may have previously remained undetected. The purpose of this systematic review was to elucidate the role of DPs in relation to brain ageing processes, by summarising current prospective and intervention studies. Nine prospective studies met the inclusion criteria for the review, seven evaluated the Mediterranean diet (MeDi), one evaluated the Alternative Healthy Eating Index-2010, and one evaluated a posteriori derived DPs. No intervention studies were eligible for inclusion in this review. There was some evidence of an association between healthy DPs and neuroimaging markers including changes within these markers over time. Consequently, it is plausible that better adherence to such DPs may positively influence brain ageing and neurodegeneration. Future studies may benefit from the use of multi-modal neuroimaging techniques, to further investigate how adherence to a DP influences brain health. The review also highlights the crucial need for further intervention studies within this research area.
Collapse
Affiliation(s)
- Rebecca F Townsend
- Centre for Public Health, Queen's University Belfast, Belfast, United Kingdom
| | - Jayne V Woodside
- Centre for Public Health, Queen's University Belfast, Belfast, United Kingdom.,Institute for Global Food Security, Queen's University Belfast, Belfast, United Kingdom
| | - Federica Prinelli
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
| | - Roisin F O'Neill
- Centre for Public Health, Queen's University Belfast, Belfast, United Kingdom
| | - Claire T McEvoy
- Centre for Public Health, Queen's University Belfast, Belfast, United Kingdom.,Institute for Global Food Security, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
16
|
Li TR, Dong QY, Jiang XY, Kang GX, Li X, Xie YY, Jiang JH, Han Y. Exploring brain glucose metabolic patterns in cognitively normal adults at risk of Alzheimer's disease: A cross-validation study with Chinese and ADNI cohorts. Neuroimage Clin 2021; 33:102900. [PMID: 34864286 PMCID: PMC8648808 DOI: 10.1016/j.nicl.2021.102900] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/17/2021] [Accepted: 11/24/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Disease-related metabolic brain patterns have been verified for a variety of neurodegenerative diseases including Alzheimer's disease (AD). This study aimed to explore and validate the pattern derived from cognitively normal controls (NCs) in the Alzheimer's continuum. METHODS This study was based on two cohorts; one from the Alzheimer's Disease Neuroimaging Initiative (ADNI) and the other from the Sino Longitudinal Study on Cognitive Decline (SILCODE). Each subject underwent [18F]fluoro-2-deoxyglucose positron emission tomography (PET) and [18F]florbetapir-PET imaging. Participants were binary-grouped based on β-amyloid (Aβ) status, and the positivity was defined as Aβ+. Voxel-based scaled subprofile model/principal component analysis (SSM/PCA) was used to generate the "at-risk AD-related metabolic pattern (ARADRP)" for NCs. The pattern expression score was obtained and compared between the groups, and receiver operating characteristic curves were drawn. Notably, we conducted cross-validation to verify the robustness and correlation analyses to explore the relationships between the score and AD-related pathological biomarkers. RESULTS Forty-eight Aβ+ NCs and 48 Aβ- NCs were included in the ADNI cohort, and 25 Aβ+ NCs and 30 Aβ- NCs were included in the SILCODE cohort. The ARADRPs were identified from the combined cohorts and the two separate cohorts, characterized by relatively lower regional loadings in the posterior parts of the precuneus, posterior cingulate, and regions of the temporal gyrus, as well as relatively higher values in the superior/middle frontal gyrus and other areas. Patterns identified from the two separate cohorts showed some regional differences, including the temporal gyrus, basal ganglia regions, anterior parts of the precuneus, and middle cingulate. Cross-validation suggested that the pattern expression score was significantly higher in the Aβ+ group of both cohorts (p < 0.01), and contributed to the diagnosis of Aβ+ NCs (with area under the curve values of 0.696-0.815). The correlation analysis revealed that the score was related to tau pathology measured in cerebrospinal fluid (p-tau: p < 0.02; t-tau: p < 0.03), but not Aβ pathology assessed with [18F]florbetapir-PET (p > 0.23). CONCLUSIONS ARADRP exists for NCs, and the acquired pattern expression score shows a certain ability to discriminate Aβ+ NCs from Aβ- NCs. The SSM/PCA method is expected to be helpful in the ultra-early diagnosis of AD in clinical practice.
Collapse
Affiliation(s)
- Tao-Ran Li
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China.
| | - Qiu-Yue Dong
- Key Laboratory of Specialty Fiber Optics and Optical Access Networks, Joint International Research Laboratory of Specialty Fiber Optics and Advanced Communication, School of Information and Communication Engineering, Shanghai University, Shanghai 200444, China.
| | - Xue-Yan Jiang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; School of Biomedical Engineering, Hainan University, Haikou 570228, China.
| | - Gui-Xia Kang
- School of Information and Communication Engineering, Beijing University of Posts and Telecommunications, Beijing 100876, China
| | - Xin Li
- School of Electrical Engineering, Yanshan University, Qinhuangdao 066004, China; Measurement Technology and Instrumentation Key Lab of Hebei Province, Qinhuangdao 066004, China
| | - Yun-Yan Xie
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China.
| | - Jie-Hui Jiang
- Key Laboratory of Specialty Fiber Optics and Optical Access Networks, Joint International Research Laboratory of Specialty Fiber Optics and Advanced Communication, School of Information and Communication Engineering, Shanghai University, Shanghai 200444, China.
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; School of Biomedical Engineering, Hainan University, Haikou 570228, China; Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing 100053, China; National Clinical Research Center for Geriatric Diseases, Beijing 100053, China.
| |
Collapse
|
17
|
Siervo M, Shannon OM, Llewellyn DJ, Stephan BC, Fontana L. Mediterranean diet and cognitive function: From methodology to mechanisms of action. Free Radic Biol Med 2021; 176:105-117. [PMID: 34562607 DOI: 10.1016/j.freeradbiomed.2021.09.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 12/12/2022]
Abstract
The traditional Mediterranean diet (MedDiet), rich in minimally processed plant foods and fish, has been widely recognized to be one of the healthiest diets. Data from multiple randomized clinical trials have demonstrated its powerful effect against oxidative stress, inflammation and the development and progression of cardiovascular disease, type 2 diabetes, and other metabolic conditions that play a crucial role in the pathogenesis of neurodegenerative diseases. The protecting effects of the MedDiet against cognitive decline have been investigated in several observational and experimental studies. Data from observational studies suggest that the MedDiet may represent an effective dietary strategy for the early prevention of dementia, although these findings require further substantiation in clinical trials which have so far produced inconclusive results. Moreover, as we discuss in this review, accumulating data emphasizes the importance of: 1) maintaining an optimal nutritional and metabolic status for the promotion of healthy cognitive aging, and 2) implementing cognition-sparing dietary and lifestyle interventions during early time-sensitive windows before the pathological cascades turn into an irreversible state. In summary, components of the MedDiet pattern, such as essential fatty acids, polyphenols and vitamins, have been associated with reduced oxidative stress and the current evidence from observational studies seems to assign to the MedDiet a beneficial role in promoting brain health; however, results from clinical trials have been inconsistent. While we advocate for longitudinal analyses and for larger and longer clinical trials to be conducted, we assert our interim support to the use of the MedDiet as a protective dietary intervention for cognitive function based on its proven cardiovascular and metabolic benefits.
Collapse
Affiliation(s)
- Mario Siervo
- School of Life Sciences, The University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK.
| | - Oliver M Shannon
- Human Nutrition Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - David J Llewellyn
- University of Exeter Medical School, Exeter, UK; Alan Turing Institute, London, UK
| | - Blossom Cm Stephan
- Institute of Mental Health, The University of Nottingham Medical School, Nottingham, UK
| | - Luigi Fontana
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Clinical and Experimental Sciences, Brescia University School of Medicine, Brescia, Italy
| |
Collapse
|
18
|
Alzheimer's Disease, Mild Cognitive Impairment and Mediterranean Diet. A Systematic Review and Dose-Response Meta-Analysis. J Clin Med 2021; 10:jcm10204642. [PMID: 34682764 PMCID: PMC8537524 DOI: 10.3390/jcm10204642] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 11/18/2022] Open
Abstract
Alzheimer’s Disease (AD) is a pathology with increasing prevalence in the context of a more long-lived society and it is the first cause of dementia in western countries. It is important to investigate factors that can be protective and may influence its development, in order to act on them trying to reduce AD incidence and its progression. The aim of this study was to conduct a systematic review and meta-analysis to determine the effects of a higher adherence to Mediterranean diet (MD) on Mild Cognitive Impairment (MCI) and AD. A literature search in PubMed, The Cochrane Library Plus and Scopus was conducted, selecting articles that analyzed associations between MD adherence and AD biomarkers (Volumetry assessed by MRI and betamiloide and Tau deposits by PET); cognitive performance in patients at risk or presenting MCI and AD; and incidence or progression from MCI to AD. Out of the 589 studies screened, 22 studies met eligibility criteria for the systematic review and qualitative synthesis. Finally, 11 studies were included in the meta-analysis (12,458 participants). Higher adherence to MD was associated with a significantly lower risk of MCI (RR = 0.91, 95%CI = 0.85–0.97) and lower risk of AD (RR = 0.89, 95% CI = 0.84–0.93). Our results enhance the importance of taking health-promoting lifestyle measures like following Mediterranean dietary patterns in order to reduce AD risk.
Collapse
|
19
|
Ballarini T, Melo van Lent D, Brunner J, Schröder A, Wolfsgruber S, Altenstein S, Brosseron F, Buerger K, Dechent P, Dobisch L, Duzel E, Ertl-Wagner B, Fliessbach K, Freiesleben SD, Frommann I, Glanz W, Hauser D, Haynes JD, Heneka MT, Janowitz D, Kilimann I, Laske C, Maier F, Metzger CD, Munk M, Perneczky R, Peters O, Priller J, Ramirez A, Rauchmann B, Roy N, Scheffler K, Schneider A, Spottke A, Spruth EJ, Teipel SJ, Vukovich R, Wiltfang J, Jessen F, Wagner M. Mediterranean Diet, Alzheimer Disease Biomarkers and Brain Atrophy in Old Age. Neurology 2021; 96:e2920-e2932. [PMID: 33952652 PMCID: PMC8253566 DOI: 10.1212/wnl.0000000000012067] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 03/15/2021] [Indexed: 02/01/2023] Open
Abstract
OBJECTIVE To determine whether following a Mediterranean-like diet (MeDi) relates to cognitive functions and in vivo biomarkers for Alzheimer disease (AD), we analyzed cross-sectional data from the German DZNE-Longitudinal Cognitive Impairment and Dementia Study. METHOD: The sample (n=512, mean age: 69.5±5.9 years) included 169 cognitively normal participants and subjects at higher AD risk (53 with relatives with AD, 209 with subjective cognitive decline, and 81 with mild cognitive impairment). We defined MeDi adherence based on the Food Frequency Questionnaire. Brain volume outcomes were generated via voxel-based morphometry on T1-MRI and cognitive performance with an extensive neuropsychological battery. AD-related biomarkers (Aβ42/40 ratio, pTau181) in cerebrospinal fluid were assessed in n=226 individuals. We analyzed the associations between MeDi and the outcomes with linear regression models controlling for several covariates. Additionally, we applied hypothesis-driven mediation and moderation analysis. RESULTS Higher MeDi adherence related to larger mediotemporal gray matter volume (p<0.05 FWE corrected), better memory (β±SE = 0.03 ± 0.02; p=0.038), and less amyloid (Aβ42/40 ratio, β±SE = 0.003 ± 0.001; p=0.008) and pTau181 pathology (β±SE = -1.96±0.68; p=0.004). Mediotemporal volume mediated the association between MeDi and memory (40% indirect mediation). Finally, MeDi favorably moderated the associations between Aβ42/40 ratio, pTau181 and mediotemporal atrophy. Results were consistent correcting for ApoE-ε4 status. CONCLUSION Our findings corroborate the view of MeDi as a protective factor against memory decline and mediotemporal atrophy. Importantly, they suggest that these associations might be explained by a decrease of amyloidosis and tau-pathology. Longitudinal and dietary intervention studies should further examine this conjecture and its treatment implications.
Collapse
Affiliation(s)
- Tommaso Ballarini
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Debora Melo van Lent
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- University of Texas Health Science Center at San Antonio: San Antonio, TX, US
| | - Julia Brunner
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Alina Schröder
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Steffen Wolfsgruber
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department of Neurodegeneration and Geriatric Psychiatry, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Slawek Altenstein
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Frederic Brosseron
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department of Neurodegeneration and Geriatric Psychiatry, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Katharina Buerger
- German Center for Neurodegenerative Diseases (DZNE, Munich), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-LynenStrasse 17, 81377 Munich, Germany
| | - Peter Dechent
- MR-Research in Neurology and Psychiatry, Georg-AugustUniversity Göttingen, Germany
| | - Laura Dobisch
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Emrah Duzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Birgit Ertl-Wagner
- Institute for Clinical Radiology, Ludwig-MaximiliansUniversity, Marchioninistr. 15, 81377 Munich
| | - Klaus Fliessbach
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department of Neurodegeneration and Geriatric Psychiatry, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Silka Dawn Freiesleben
- Charité Universitätsmedizin Berlin, Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Ingo Frommann
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Dietmar Hauser
- Charité Universitätsmedizin Berlin, Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - John Dylan Haynes
- Bernstein Center for Computational Neuroscience, Charité Universitätsmedizin, Berlin, Germany
| | - Michael T Heneka
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department of Neurodegeneration and Geriatric Psychiatry, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Str. 20, 18147 Rostock
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Franziska Maier
- Department of Psychiatry, University of Cologne, Medical Faculty, Kerpener Strasse 62, 50924 Cologne, Germany
| | - Coraline Danielle Metzger
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Matthias Munk
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE, Munich), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London, UK
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Charité Universitätsmedizin Berlin, Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Alfredo Ramirez
- Department of Psychiatry, University of Cologne, Medical Faculty, Kerpener Strasse 62, 50924 Cologne, Germany
| | - Boris Rauchmann
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Nina Roy
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Klaus Scheffler
- Department for Biomedical Magnetic Resonance, University of Tübingen, 72076 Tübingen, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department of Neurodegeneration and Geriatric Psychiatry, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department of Neurology, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Eike Jakob Spruth
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Stefan J Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Str. 20, 18147 Rostock
| | - Ruth Vukovich
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Von-Siebold-Str. 5, 37075 Goettingen
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Von-Siebold-Str. 5, 37075 Goettingen
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED)
- Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department of Psychiatry, University of Cologne, Medical Faculty, Kerpener Strasse 62, 50924 Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931Köln, Germany
| | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department of Neurodegeneration and Geriatric Psychiatry, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| |
Collapse
|
20
|
Poddar MK, Banerjee S, Chakraborty A, Dutta D. Metabolic disorder in Alzheimer's disease. Metab Brain Dis 2021; 36:781-813. [PMID: 33638805 DOI: 10.1007/s11011-021-00673-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/14/2021] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD), a well known aging-induced neurodegenerative disease is related to amyloid proteinopathy. This proteinopathy occurs due to abnormalities in protein folding, structure and thereby its function in cells. The root cause of such kind of proteinopathy and its related neurodegeneration is a disorder in metabolism, rather metabolomics of the major as well as minor nutrients. Metabolomics is the most relevant "omics" platform that offers a great potential for the diagnosis and prognosis of neurodegenerative diseases as an individual's metabolome. In recent years, the research on such kinds of neurodegenerative diseases, especially aging-related disorders is broadened its scope towards metabolic function. Different neurotransmitter metabolisms are also involved with AD and its associated neurodegeneration. The genetic and epigenetic backgrounds are also noteworthy. In this review, the physiological changes of AD in relation to its corresponding biochemical, genetic and epigenetic involvements including its (AD) therapeutic aspects are discussed.
Collapse
Affiliation(s)
- Mrinal K Poddar
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata, 700032, India.
| | - Soumyabrata Banerjee
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata, 700032, India
- Departrment of Psychology, Neuroscience Program, Field Neurosciences Institute Research Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Apala Chakraborty
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata, 700032, India
| | - Debasmita Dutta
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata, 700032, India
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND, 58102, USA
| |
Collapse
|
21
|
Samuelsson J, Kern S, Zetterberg H, Blennow K, Rothenberg E, Wallengren O, Skoog I, Zettergren A. A Western-style dietary pattern is associated with cerebrospinal fluid biomarker levels for preclinical Alzheimer's disease-A population-based cross-sectional study among 70-year-olds. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12183. [PMID: 34027029 PMCID: PMC8129853 DOI: 10.1002/trc2.12183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 03/17/2021] [Accepted: 04/20/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND Diet may be a modifiable factor for reducing the risk of Alzheimer's disease (AD). Western-style dietary patterns are considered to increase the risk, whereas Mediterranean-style dietary patterns are considered to reduce the risk. An association between diet and AD-related biomarkers have been suggested, but studies are limited. AIM To investigate potential relations between dietary patterns and cerebrospinal fluid (CSF) biomarkers for AD among dementia-free older adults. METHODS Data were derived from the population-based Gothenburg H70 Birth Cohort Studies, Sweden. A total of 269 dementia-free 70-year-olds with dietary and cerebrospinal fluid (CSF) amyloid beta (Aβ42 and Aβ40), total tau (t-tau), and phosphorylated tau (p-tau) data were investigated. Dietary intake was determined by the diet history method, and four dietary patterns were derived by principal component analysis. A Western dietary pattern, a Mediterranean/prudent dietary pattern, a high-protein and alcohol pattern, and a high-total and saturated fat pattern. Logistic regression models, with CSF biomarker pathology (yes/no) as dependent variables, and linear regression models with continuous CSF biomarker levels as dependent variables were performed. The analyses were adjusted for sex, energy intake, body mass index (BMI), educational level, and physical activity level. RESULTS The odds ratio for having total tau pathology (odds ratio [OR] 1.43; 95% confidence interval [CI] 1.02 to 2.01) and preclinical AD (Aβ42 and tau pathology; OR 1.79; 95% CI 1.03 to 3.10) was higher among those with a higher adherence to a Western dietary pattern. There were no other associations between the dietary patterns and CSF biomarkers that remained significant in both unadjusted and adjusted models. DISCUSSION Our findings suggest that higher adherence to a Western dietary pattern may be associated with pathological levels of AD biomarkers in the preclinical phase of AD. These findings can be added to the increasing amount of evidence linking diet with AD and may be useful for future intervention studies investigating dietary intake in relation to AD.
Collapse
Affiliation(s)
- Jessica Samuelsson
- Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, Sahlgrenska Academy, Centre for Ageing and Health (AGECAP) at the University of GothenburgGothenburgSweden
| | - Silke Kern
- Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, Sahlgrenska Academy, Centre for Ageing and Health (AGECAP) at the University of GothenburgGothenburgSweden
| | - Henrik Zetterberg
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska Academy at the University of GothenburgGothenburgSweden
- UK Dementia Research Institute at UCLLondonUK
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
| | - Kaj Blennow
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska Academy at the University of GothenburgGothenburgSweden
| | | | - Ola Wallengren
- Clinical Nutrition UnitSahlgrenska University HospitalGothenburgSweden
| | - Ingmar Skoog
- Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, Sahlgrenska Academy, Centre for Ageing and Health (AGECAP) at the University of GothenburgGothenburgSweden
| | - Anna Zettergren
- Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, Sahlgrenska Academy, Centre for Ageing and Health (AGECAP) at the University of GothenburgGothenburgSweden
| |
Collapse
|
22
|
Wesselman LMP, van Lent DM, Schröder A, van de Rest O, Peters O, Menne F, Fuentes M, Priller J, Spruth EJ, Altenstein S, Schneider A, Fließbach K, Roeske S, Wolfsgruber S, Kleineidam L, Spottke A, Pross V, Wiltfang J, Vukovich R, Schild AK, Düzel E, Metzger CD, Glanz W, Buerger K, Janowitz D, Perneczky R, Tatò M, Teipel S, Kilimann I, Laske C, Buchmann M, Ramirez A, Sikkes SAM, Jessen F, van der Flier WM, Wagner M. Dietary patterns are related to cognitive functioning in elderly enriched with individuals at increased risk for Alzheimer's disease. Eur J Nutr 2021; 60:849-860. [PMID: 32472387 PMCID: PMC7900077 DOI: 10.1007/s00394-020-02257-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 04/22/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE To investigate cross-sectional associations between dietary patterns and cognitive functioning in elderly free of dementia. METHODS Data of 389 participants from the German DELCODE study (52% female, 69 ± 6 years, mean Mini Mental State Score 29 ± 1) were included. The sample was enriched with elderly at increased risk for Alzheimer's disease (AD) by including participants with subjective cognitive decline, mild cognitive impairment (MCI) and siblings of AD patients. Mediterranean and MIND diets were derived from 148 Food Frequency Questionnaire items, and data-driven patterns by principal component analysis (PCA) of 39 food groups. Associations between dietary patterns and five cognitive domain scores were analyzed with linear regression analyses adjusted for demographics (model 1), and additionally for energy intake, BMI, other lifestyle variables and APOe4-status (model 2). For PCA-derived dietary components, final model 3 included all other dietary components. RESULTS In fully adjusted models, adherence to Mediterranean and MIND diet was associated with better memory. The 'alcoholic beverages' PCA component was positively associated with most cognitive domains. Exclusion of MCI subjects (n = 60) revealed that Mediterranean and MIND diet were also related to language functions; associations with the alcoholic beverages component were attenuated, but most remained significant. CONCLUSION In line with data from elderly population samples, Mediterranean and MIND diet and some data-derived dietary patterns were related to memory and language function. Longitudinal data are needed to draw conclusions on the putative effect of nutrition on the rate of cognitive decline, and on the potential of dietary interventions in groups at increased risk for AD.
Collapse
Affiliation(s)
- L. M. P. Wesselman
- German Center for Neurodegenerative Disorders (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - D. Melo van Lent
- German Center for Neurodegenerative Disorders (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- The Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, UT Health, San Antonio, TX USA
- Department of Neurology, Boston University, Boston, MA USA
- The Framingham Heart Study, Framingham, MA USA
| | - A. Schröder
- German Center for Neurodegenerative Disorders (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - O. van de Rest
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, The Netherlands
| | - O. Peters
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Psychiatry and Psychotherapy, Hindenburgdamm 30, 12203 Berlin, Germany
| | - F. Menne
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Psychiatry and Psychotherapy, Hindenburgdamm 30, 12203 Berlin, Germany
| | - M. Fuentes
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Psychiatry and Psychotherapy, Hindenburgdamm 30, 12203 Berlin, Germany
| | - J. Priller
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - E. J. Spruth
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - S. Altenstein
- Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - A. Schneider
- German Center for Neurodegenerative Disorders (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - K. Fließbach
- German Center for Neurodegenerative Disorders (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - S. Roeske
- German Center for Neurodegenerative Disorders (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - S. Wolfsgruber
- German Center for Neurodegenerative Disorders (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - L. Kleineidam
- German Center for Neurodegenerative Disorders (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - A. Spottke
- German Center for Neurodegenerative Disorders (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department of Neurology, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - V. Pross
- Study Center Bonn, Medical Faculty, Venusberg-Campus 1, 53127 Bonn, Germany
| | - J. Wiltfang
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Von-Siebold-Str. 5, 37075 Goettingen , Germany
| | - R. Vukovich
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Von-Siebold-Str. 5, 37075 Goettingen , Germany
| | - A. K. Schild
- German Center for Neurodegenerative Disorders (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department of Psychiatry, University of Cologne, Medical Faculty, Kerpener Strasse 62, 50924 Cologne, Germany
| | - E. Düzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-Von-Guericke University, Magdeburg, Germany
| | - C. D. Metzger
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-Von-Guericke University, Magdeburg, Germany
- Department of Psychiatry and Psychotherapy, Otto-Von-Guericke University, Magdeburg, Germany
| | - W. Glanz
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - K. Buerger
- German Center for Neurodegenerative Diseases (DZNE, Munich), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - D. Janowitz
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - R. Perneczky
- German Center for Neurodegenerative Diseases (DZNE, Munich), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London, UK
| | - M. Tatò
- German Center for Neurodegenerative Diseases (DZNE, Munich), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - S. Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | - I. Kilimann
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | - C. Laske
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - M. Buchmann
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - A. Ramirez
- Department of Psychiatry, University of Cologne, Medical Faculty, Kerpener Strasse 62, 50924 Cologne, Germany
| | - S. A. M. Sikkes
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Clinical Developmental Psychology and Clinical Neuropsychology, Faculty of Behavioural and Movement Sciences (FGB), Vrije University Amsterdam, Amsterdam, The Netherlands
| | - F. Jessen
- German Center for Neurodegenerative Disorders (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department of Psychiatry, University of Cologne, Medical Faculty, Kerpener Strasse 62, 50924 Cologne, Germany
| | - W. M. van der Flier
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Epidemiology and Biostatistics, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - M. Wagner
- German Center for Neurodegenerative Disorders (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| |
Collapse
|
23
|
Vidoni ED, Morris JK, Watts A, Perry M, Clutton J, Van Sciver A, Kamat AS, Mahnken J, Hunt SL, Townley R, Honea R, Shaw AR, Johnson DK, Vacek J, Burns JM. Effect of aerobic exercise on amyloid accumulation in preclinical Alzheimer's: A 1-year randomized controlled trial. PLoS One 2021; 16:e0244893. [PMID: 33444359 PMCID: PMC7808620 DOI: 10.1371/journal.pone.0244893] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Our goal was to investigate the role of physical exercise to protect brain health as we age, including the potential to mitigate Alzheimer's-related pathology. We assessed the effect of 52 weeks of a supervised aerobic exercise program on amyloid accumulation, cognitive performance, and brain volume in cognitively normal older adults with elevated and sub-threshold levels of cerebral amyloid as measured by amyloid PET imaging. METHODS AND FINDINGS This 52-week randomized controlled trial compared the effects of 150 minutes per week of aerobic exercise vs. education control intervention. A total of 117 underactive older adults (mean age 72.9 [7.7]) without evidence of cognitive impairment, with elevated (n = 79) or subthreshold (n = 38) levels of cerebral amyloid were randomized, and 110 participants completed the study. Exercise was conducted with supervision and monitoring by trained exercise specialists. We conducted 18F-AV45 PET imaging of cerebral amyloid and anatomical MRI for whole brain and hippocampal volume at baseline and Week 52 follow-up to index brain health. Neuropsychological tests were conducted at baseline, Week 26, and Week 52 to assess executive function, verbal memory, and visuospatial cognitive domains. Cardiorespiratory fitness testing was performed at baseline and Week 52 to assess response to exercise. The aerobic exercise group significantly improved cardiorespiratory fitness (11% vs. 1% in the control group) but there were no differences in change measures of amyloid, brain volume, or cognitive performance compared to control. CONCLUSIONS Aerobic exercise was not associated with reduced amyloid accumulation in cognitively normal older adults with cerebral amyloid. In spite of strong systemic cardiorespiratory effects of the intervention, the observed lack of cognitive or brain structure benefits suggests brain benefits of exercise reported in other studies are likely to be related to non-amyloid effects. TRIAL REGISTRATION NCT02000583; ClinicalTrials.gov.
Collapse
Affiliation(s)
- Eric D. Vidoni
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
| | - Jill K. Morris
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
| | - Amber Watts
- Department of Psychology, University of Kansas, Lawrence, KS, United States of America
| | - Mark Perry
- Department of Radiology, University of Kansas Health System, Kansas City, KS, United States of America
| | - Jon Clutton
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
| | - Angela Van Sciver
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
| | - Ashwini S. Kamat
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
| | - Jonathan Mahnken
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Suzanne L. Hunt
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Ryan Townley
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
| | - Robyn Honea
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
| | - Ashley R. Shaw
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
| | - David K. Johnson
- Department of Neurology, University of California–Davis, Sacramento, CA, United States of America
| | - James Vacek
- Department of Cardiovascular Medicine, University of Kansas Health System, Kansas City, KS, United States of America
| | - Jeffrey M. Burns
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
| |
Collapse
|
24
|
van Aalst J, Jennen L, Demyttenaere K, Sunaert S, Koole M, Ceccarini J, Van Laere K. Twelve-Week Yoga vs. Aerobic Cycling Initiation in Sedentary Healthy Subjects: A Behavioral and Multiparametric Interventional PET/MR Study. Front Psychiatry 2021; 12:739356. [PMID: 34733191 PMCID: PMC8558251 DOI: 10.3389/fpsyt.2021.739356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/16/2021] [Indexed: 11/21/2022] Open
Abstract
Interventional yoga studies with an active control group remain scarce and are important to clarify the underlying neurobiology. We conducted an interventional study in healthy controls using simultaneous positron emission tomography/magnetic resonance (PET/MR) imaging and psychometric scales. Thirty healthy, female volunteers (28.4 ± 8.4 years) participated and were randomly assigned to a 12-week yoga or indoor cycling intervention. Before and after the intervention, [18F]FDG and [11C]UCB-J PET was performed on a simultaneous GE Signa PET/MR with volumetric imaging. Psychometric scales were evaluated on affect, mindfulness, stress, worrying, self-compassion, and interoceptive awareness. Yoga subjects scored higher on interoceptive awareness compared to baseline (p < 0.001). Cognitive (P = 0.009) and overall cognitive functioning (P = 0.01) improved after the yoga intervention compared to the cycling group. We did not observe significant differences in glucose metabolism, synaptic density, or gray matter (GM) volume. The indoor cycling group did not show changes in psychometric variables, but significant increases in relative glucose metabolism were observed in the parahippocampal/fusiform gyrus and cerebellum (P < 0.001). In conclusion, 12 weeks of yoga practice has significant effects on interoceptive awareness and perceived cognitive function in starters. Longer interventions and/or higher frequency of yoga practice may be needed to detect cerebral metabolic and/or morphologic effects on the macroscopic level.
Collapse
Affiliation(s)
- June van Aalst
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Lise Jennen
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Koen Demyttenaere
- Research Group Psychiatry, Neurosciences, University Psychiatric Center KU Leuven, Leuven, Belgium.,Adult Psychiatry, University Hospitals Leuven, Leuven, Belgium
| | - Stefan Sunaert
- Translational MRI, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.,Radiology, University Hospitals Leuven, Leuven, Belgium
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Jenny Ceccarini
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.,Division of Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
25
|
Specific nutrient patterns are associated with higher structural brain integrity in dementia-free older adults. Neuroimage 2019; 199:281-288. [DOI: 10.1016/j.neuroimage.2019.05.066] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/21/2019] [Accepted: 05/26/2019] [Indexed: 01/19/2023] Open
|
26
|
Nutrients in the Prevention of Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9874159. [PMID: 31565158 PMCID: PMC6746160 DOI: 10.1155/2019/9874159] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/02/2019] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a disease caused by the complex interaction of multiple mechanisms, some of which are still not fully understood. To date, pharmacological treatments and supplementation of individual nutrients have been poorly effective in terms of the prevention and treatment of AD, while alternative strategies based on multimodal approaches (diet, exercise, and cognitive training) seem to be more promising. In this context, the focus on dietary patterns rather than on single food components could be more useful in preventing or counteracting the pathological processes typical of AD, thanks to the potential synergistic effects of various nutrients (neuronutrients). The aim of this narrative review is to summarize the currently existing preclinical and clinical evidence regarding the Mediterranean diet (MeDi), the Dietary Approaches to Stop Hypertension (DASH) diet, and the Mediterranean-DASH Intervention for Neurodegenerative Delay (MIND) diet, which are three dietary patterns with well-known anti-inflammatory and antioxidant properties. Recently, they have been related to brain protection and AD prevention, perhaps thanks to their high content of neuroprotective bioactive compounds. Similarly, intermittent fasting (IF) or calorie restriction (CR) is emerging as interesting approaches that seem to promote hippocampal neurogenesis, activate adaptive stress response systems, and enhance neuronal plasticity, thus leading to motor and cognitive improvements in animal models of AD and hopefully also in human beings.
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW The purposes of this review were to examine literature published over the last 5 years and to evaluate the role of nutrition in cognitive function and brain ageing, focussing on the Mediterranean diet (MeDi), Dietary Approaches to Stop Hypertension (DASH), and Mediterranean-DASH Intervention for Neurodegenerative Delay (MIND) diets. RECENT FINDINGS Results suggest that higher adherence to a healthy dietary pattern is associated with preservation of brain structure and function as well as slower cognitive decline, with the MIND diet substantially slowing cognitive decline, over and above the MeDi and DASH diets. Whilst results to-date suggest adherence to a healthy diet, such as the MeDi, DASH, or MIND, is an important modifiable risk factor in the quest to develop strategies aimed at increasing likelihood of healthy brain ageing, further work is required to develop dietary guidelines with the greatest potential benefit for public health; a research topic of increasing importance as the world's population ages.
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW We reviewed the most recent literature examining the associations between the Mediterranean-style diet (MD), neurodegenerative diseases, and markers and mechanisms of neurodegeneration. RECENT FINDINGS Most, but not all, epidemiologic studies report a protective association between MD adherence, cognitive impairment, and brain health. Data from clinical trials supporting these observational findings are also emerging. Limited evidence suggests that MD adherence may be protective for Parkinson's disease risk. Mechanistically, plant polyphenols may activate similar molecular pathways as caloric restriction diets, which helps explain the neuroprotective properties of the MD. Evidence for cognitive disorders is abundant, but there is a dearth of literature for other neurodegenerative disorders and for markers of neurodegeneration. Further research is needed to elucidate the protective role of MD on neurodegeneration, the most salient components of the MD, and the most sensitive time periods over the lifecourse at which the MD may exert its effects.
Collapse
Affiliation(s)
- Hannah Gardener
- Department of Neurology, Miller School of Medicine, and Evelyn F. McKnight Brain Institute, University of Miami, 1120 NW 14th Street, 13th Floor, Miami, FL, 33136, USA.
| | - Michelle R Caunca
- Department of Neurology, Miller School of Medicine, and Evelyn F. McKnight Brain Institute, University of Miami, 1120 NW 14th Street, 13th Floor, Miami, FL, 33136, USA.,Division of Epidemiology and Population Health Sciences, Department of Public Health Sciences, Miller School of Medicine, University of Miami, 1120 NW 14th Street, 1007B, Miami, FL, 33136, USA
| |
Collapse
|
29
|
Sánchez-Muniz FJ, Macho-González A, Garcimartín A, Santos-López JA, Benedí J, Bastida S, González-Muñoz MJ. The Nutritional Components of Beer and Its Relationship with Neurodegeneration and Alzheimer's Disease. Nutrients 2019; 11:nu11071558. [PMID: 31295866 PMCID: PMC6682961 DOI: 10.3390/nu11071558] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/04/2019] [Accepted: 07/08/2019] [Indexed: 02/07/2023] Open
Abstract
The prevalence of degenerative diseases has risen in western countries. Growing evidence suggests that demenia and other cognition affectations are associated with ambient factors including specific nutrients, food ingredients or specific dietary patterns. Mediterranean diet adherence has been associated with various health benefits and decreased risk of many diseases, including neurodegenerative disorders. Beer, as part of this protective diet, contains compounds such as silicon and hops that could play a major role in preventing brain disorders. In this review, different topics regarding Mediterranean diet, beer and the consumption of their main compounds and their relation to neurological health have been addressed. Taking into account published results from our group and other studies, the hypothesis linking aluminum intoxication with dementia and/or Alzheimer’s disease and the potential role of regular beer has also been considered. Beer, in spite of its alcohol content, may have some health benefits; nonetheless, its consumption is not adequate for all subjects. Thus, this review analyzed some promising results of non-alcoholic beer on several mechanisms engaged in neurodegeneration such as inflammation, oxidation, and cholinesterase activity, and their contribution to the behavioral modifications induced by aluminum intoxication. The review ends by giving conclusions and suggesting future topics of research related to moderate beer consumption and/or the consumption of its major compounds as a potential instrument for protecting against neurodegenerative disease progression and the need to develop nutrigenetic and nutrigenomic studies in aged people and animal models.
Collapse
Affiliation(s)
- Francisco José Sánchez-Muniz
- Departamento de Nutrición y Ciencia de los Alimentos, Facultad de Farmacia. Universidad Complutense de Madrid, 28040 Madrid, Spain.
- AFUSAN Research Group. Universidad Complutense de Madrid and Instituto de Investigación Sanitaria from Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain.
| | - Adrián Macho-González
- Departamento de Nutrición y Ciencia de los Alimentos, Facultad de Farmacia. Universidad Complutense de Madrid, 28040 Madrid, Spain
- AFUSAN Research Group. Universidad Complutense de Madrid and Instituto de Investigación Sanitaria from Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Alba Garcimartín
- AFUSAN Research Group. Universidad Complutense de Madrid and Instituto de Investigación Sanitaria from Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia. Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Jorge Arturo Santos-López
- AFUSAN Research Group. Universidad Complutense de Madrid and Instituto de Investigación Sanitaria from Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia. Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Juana Benedí
- AFUSAN Research Group. Universidad Complutense de Madrid and Instituto de Investigación Sanitaria from Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia. Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Sara Bastida
- Departamento de Nutrición y Ciencia de los Alimentos, Facultad de Farmacia. Universidad Complutense de Madrid, 28040 Madrid, Spain
- AFUSAN Research Group. Universidad Complutense de Madrid and Instituto de Investigación Sanitaria from Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - María José González-Muñoz
- AFUSAN Research Group. Universidad Complutense de Madrid and Instituto de Investigación Sanitaria from Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Departamento de Ciencias Biomédicas, Unidad Docente de Toxicología, Facultad de Farmacia, Universidad de Alcalá, 28805 Alcalá de Henares, Spain
| |
Collapse
|
30
|
Meyer PF, McSweeney M, Gonneaud J, Villeneuve S. AD molecular: PET amyloid imaging across the Alzheimer's disease spectrum: From disease mechanisms to prevention. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 165:63-106. [PMID: 31481172 DOI: 10.1016/bs.pmbts.2019.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The advent of amyloid-beta (Aβ) positron emission tomography (PET) imaging has transformed the field of Alzheimer's disease (AD) by enabling the quantification of cortical Aβ accumulation and propagation in vivo. This revolutionary tool has made it possible to measure direct associations between Aβ and other AD biomarkers, to identify factors that influence Aβ accumulation and to redefine entry criteria into clinical trials as well as measure drug target engagement. This chapter summarizes the main findings on the associations of Aβ with other biomarkers of disease progression across the AD spectrum. It discusses investigations of the timing at which Aβ pathology starts to accumulate, demonstrates the clinical utility of Aβ PET imaging and discusses some ethical implications. Finally, it presents genetic and potentially modifiable lifestyle factors that might influence Aβ accumulation and therefore be targets for AD prevention.
Collapse
Affiliation(s)
- Pierre-François Meyer
- Centre for Studies on the Prevention of Alzheimer's Disease, Douglas Mental Health University Institute, Montréal, Canada; McGill University, Montréal, Canada
| | - Melissa McSweeney
- Centre for Studies on the Prevention of Alzheimer's Disease, Douglas Mental Health University Institute, Montréal, Canada; McGill University, Montréal, Canada
| | - Julie Gonneaud
- Centre for Studies on the Prevention of Alzheimer's Disease, Douglas Mental Health University Institute, Montréal, Canada; McGill University, Montréal, Canada
| | - Sylvia Villeneuve
- Centre for Studies on the Prevention of Alzheimer's Disease, Douglas Mental Health University Institute, Montréal, Canada; McGill University, Montréal, Canada.
| |
Collapse
|
31
|
Hill E, Goodwill AM, Gorelik A, Szoeke C. Diet and biomarkers of Alzheimer's disease: a systematic review and meta-analysis. Neurobiol Aging 2019; 76:45-52. [DOI: 10.1016/j.neurobiolaging.2018.12.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 11/29/2018] [Accepted: 12/18/2018] [Indexed: 11/16/2022]
|
32
|
Zwilling CE, Talukdar T, Zamroziewicz MK, Barbey AK. Nutrient biomarker patterns, cognitive function, and fMRI measures of network efficiency in the aging brain. Neuroimage 2019; 188:239-251. [DOI: 10.1016/j.neuroimage.2018.12.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/28/2018] [Accepted: 12/04/2018] [Indexed: 11/26/2022] Open
|
33
|
James BD, Bennett DA. Causes and Patterns of Dementia: An Update in the Era of Redefining Alzheimer's Disease. Annu Rev Public Health 2019; 40:65-84. [PMID: 30642228 DOI: 10.1146/annurev-publhealth-040218-043758] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The burden of dementia continues to increase as the population ages, with no disease-modifying treatments available. However, dementia risk appears to be decreasing, and progress has been made in understanding its multifactorial etiology. The 2018 National Institute on Aging-Alzheimer's Association (NIA-AA) research framework for Alzheimer's disease (AD) defines AD as a biological process measured by brain pathology or biomarkers, spanning the cognitive spectrum from normality to dementia. This framework facilitates interventions in the asymptomatic space and accommodates knowledge that many additional pathologies (e.g., cerebrovascular) contribute to the Alzheimer's dementia syndrome. The framework has implications for how we think about risk factors for "AD": Many commonly accepted risk factors are not related to AD pathology and would no longer be considered risk factors for AD. They may instead be related to other pathologies or resilience to pathology. This review updates what is known about causes, risk factors, and changing patterns of dementia, addressing whether they are related to AD pathology/biomarkers, other pathologies, or resilience.
Collapse
Affiliation(s)
- Bryan D James
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois 60612, USA; .,Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois 60612, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois 60612, USA; .,Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612, USA
| |
Collapse
|
34
|
Mediterranean diet adherence and rate of cerebral Aβ-amyloid accumulation: Data from the Australian Imaging, Biomarkers and Lifestyle Study of Ageing. Transl Psychiatry 2018; 8:238. [PMID: 30375373 PMCID: PMC6207555 DOI: 10.1038/s41398-018-0293-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 07/15/2018] [Accepted: 07/24/2018] [Indexed: 12/26/2022] Open
Abstract
Accumulating research has linked Mediterranean diet (MeDi) adherence with slower cognitive decline and reduced Alzheimer's disease (AD) risk. However, no study to-date has examined the relationship between MeDi adherence and accumulation of cerebral Aβ-amyloid (Aβ; a pathological hallmark of AD) in older adults. Cognitively normal healthy control participants of the Australian Imaging, Biomarkers and Lifestyle (AIBL) Study of Ageing completed the Cancer Council of Victoria Food Frequency Questionnaire at baseline, which was used to construct a MeDi score for each participant (score range 0-9; higher score indicating higher adherence). Cerebral Aβ load was quantified by Pittsburgh Compound B positron emission tomography at baseline, 18 and 36 months: Only individuals categorised as "Aβ accumulators", and thus considered to be on the AD pathway, were included in the analysis (N = 77). The relationship between MeDi adherence, MeDi components, and change in cerebral Aβ load (baseline to 36 months) was evaluated using Generalised Linear Modelling, accounting for age, gender, education, Apolipoprotein E ε4 allele status, body mass index and total energy intake. Higher MeDi score was associated with less Aβ accumulation in our cohort (β = -0.01 ± 0.004, p = 0.0070). Of the individual MeDi score components, a high intake of fruit was associated with less accumulation of Aβ (β = -0.04 ± 0.01, p = 0.00036). Our results suggest MeDi adherence is associated with reduced cerebral AD pathology accumulation over time. When our results are considered collectively with previous data linking the MeDi to slower cognitive decline, it is apparent that MeDi adherence warrants further investigation in the quest to delay AD onset.
Collapse
|
35
|
Cattaneo G, Bartrés-Faz D, Morris TP, Sánchez JS, Macià D, Tarrero C, Tormos JM, Pascual-Leone A. The Barcelona Brain Health Initiative: A Cohort Study to Define and Promote Determinants of Brain Health. Front Aging Neurosci 2018; 10:321. [PMID: 30405394 PMCID: PMC6204574 DOI: 10.3389/fnagi.2018.00321] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 09/24/2018] [Indexed: 01/25/2023] Open
Abstract
The Barcelona Brain Health Initiative (BBHI) is an ongoing prospective longitudinal study focused on identifying determinants of brain health. The main objectives are: (i) to characterize lifestyle, cognitive, behavioral and environmental markers related to a given individual's cognitive and mental functions in middle to old age, (ii) to assess the biological determinants predictive of maintenance of brain health, and (iii) to evaluate the impact of a controlled multi-dimensional lifestyle intervention on improving and maintaining brain health. The BBHI cohort consists of >4500 healthy participants aged 40-65 years followed through online questionnaires (Phase I) assessing participants' self-perceived health and lifestyle factors in seven different domains: overall health, physical exercise, cognitive activity, sleep, nutrition, social interactions, and life purpose. In Phase II a sub-group of 1,000 individuals is undergoing detailed in-person evaluations repeated at two-yearly intervals. These evaluations will provide deep phenotyping of brain function, including medical, neurological and psychiatric examinations, assessment of physical fitness, neuropsychological assessments, structural and functional brain magnetic resonance imaging, electroencephalography and perturbation-based non-invasive brain stimulation evaluations of brain activity, as well as collection of biological samples. Finally, in Phase III a further sub-group of 500 participants will undergo a similar in-person assessment before and after a multi-dimensional intervention to optimize lifestyle habits and evaluate its effects on cognitive and brain structure and function. The intervention group will receive remote supervision through an ICT-based solution, with the support of an expert in health and lifestyle coaching strategies aimed at promoting adherence. On the other hand, the control group will not have this coaching support, and will only receive education and recommendations about healthy habits. Results of this three-part initiative shall critically contribute to a better understanding of the determinants to promote and maintain brain health over the lifespan.
Collapse
Affiliation(s)
- Gabriele Cattaneo
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Badalona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - David Bartrés-Faz
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Badalona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Departament de Medicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Timothy P. Morris
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Badalona, Spain
- Universitat Autònoma de Barcelona, Barcelona, Spain
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Javier Solana Sánchez
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Badalona, Spain
- Universitat Autònoma de Barcelona, Barcelona, Spain
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Dídac Macià
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Badalona, Spain
- Universitat Autònoma de Barcelona, Barcelona, Spain
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Clara Tarrero
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Badalona, Spain
- Universitat Autònoma de Barcelona, Barcelona, Spain
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Josep M. Tormos
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Badalona, Spain
- Universitat Autònoma de Barcelona, Barcelona, Spain
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Alvaro Pascual-Leone
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Badalona, Spain
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
36
|
Ravi SK, Narasingappa RB, Vincent B. Neuro-nutrients as anti-alzheimer's disease agents: A critical review. Crit Rev Food Sci Nutr 2018; 59:2999-3018. [PMID: 29846084 DOI: 10.1080/10408398.2018.1481012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease (AD) is characterized by a massive neuronal death causing memory loss, cognitive impairment and behavioral alteration that ultimately lead to dementia and death. AD is a multi-factorial pathology controlled by molecular events such as oxidative stress, protein aggregation, mitochondrial dysfunction and neuro inflammation. Nowadays, there is no efficient disease-modifying treatment for AD and epidemiological studies have suggested that diet and nutrition have a significant impact on the development of this disorder. Indeed, some nutrients can protect all kind of cells, including neurons. As prevention is better than cure, life style improvement, with a special emphasis on diet, should seriously be considered as an anti-AD track and intake of nutrients promoting neuronal health is the need of the hour. Diets rich in unsaturated fatty acids, polyphenols and vitamins have been shown to protect against AD, whereas saturated fatty acids-containing diets deprived of polyphenols promote the development of the disease. Thus, Mediterranean diets, mainly composed of fruits, vegetables and omega-3 fatty acids, stand as valuable, mild and preventive anti-AD agents. This review focuses on our current knowledge in the field and how one can fight this devastating neurodegenerative disorder through the simple proper modification of our life style.
Collapse
Affiliation(s)
- Sunil K Ravi
- Department of Biotechnology, College of Agriculture, University of Agriculture Sciences , Bangalore , Hassan , Karnataka , India
| | - Ramesh B Narasingappa
- Department of Biotechnology, College of Agriculture, University of Agriculture Sciences , Bangalore , Hassan , Karnataka , India
| | - Bruno Vincent
- Institute of Molecular Biosciences, Mahidol University , Nakhon Pathom , Thailand.,Centre National de la Recherche Scientifique , Paris , France
| |
Collapse
|
37
|
Vos SJ, Visser PJ. Preclinical Alzheimer’s Disease: Implications for Refinement of the Concept. J Alzheimers Dis 2018; 64:S213-S227. [DOI: 10.3233/jad-179943] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Stephanie J.B. Vos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, Netherlands
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, Netherlands
- Department of Neurology, Alzheimer Center, VU University Medical Center, Amsterdam, Netherlands
| |
Collapse
|
38
|
Dougherty RJ, Schultz SA, Kirby TK, Boots EA, Oh JM, Edwards D, Gallagher CL, Carlsson CM, Bendlin BB, Asthana S, Sager MA, Hermann BP, Christian BT, Johnson SC, Cook DB, Okonkwo OC. Moderate Physical Activity is Associated with Cerebral Glucose Metabolism in Adults at Risk for Alzheimer's Disease. J Alzheimers Dis 2018; 58:1089-1097. [PMID: 28527205 DOI: 10.3233/jad-161067] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The objective of this study was to investigate the relationship between accelerometer-measured physical activity (PA) and glucose metabolism in asymptomatic late-middle-aged adults. Ninety-three cognitively healthy late-middle-aged adults from the Wisconsin Registry for Alzheimer's Prevention participated in this cross-sectional study. They underwent 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) imaging and wore an accelerometer (ActiGraph GT3X+) to measure free-living PA. Accelerometer data yielded measures of light (LPA), moderate (MPA), and vigorous (VPA) intensity PA. FDG-PET images were scaled to the cerebellum and pons, and cerebral glucose metabolic rate was extracted from specific regions of interest (ROIs) known to be hypometabolic in AD, i.e., hippocampus, posterior cingulate, inferior temporal cortex, and angular gyrus. Regression analyses were utilized to examine the association between PA and glucose metabolism, while adjusting for potential confounds. There were associations between MPA and glucose metabolism in all ROIs examined. In contrast, LPA was not associated with glucose uptake in any ROI and VPA was only associated with hippocampal FDG uptake. Secondary analyses did not reveal associations between sedentary time and glucose metabolism in any of the ROIs. Exploratory voxel-wise analysis identified additional regions where MPA was significantly associated with glucose metabolism including the precuneus, supramarginal gyrus, amygdala, and middle frontal gyrus. These findings suggest that the intensity of PA is an important contributor to neuronal function in a late-middle-aged cohort, with MPA being the most salient. Prospective studies are necessary for fully elucidating the link between midlife engagement in PA and later life development of AD.
Collapse
Affiliation(s)
- Ryan J Dougherty
- Department of Kinesiology, University of Wisconsin School of Education, Madison, WI, USA.,Wisconsin Alzheimer's Disease Research Center
| | - Stephanie A Schultz
- Wisconsin Alzheimer's Disease Research Center.,Geriatric Research Education and Clinical Center, William S. Middleton Memorial VA Hospital, Madison, WI, USA
| | - Taylor K Kirby
- Wisconsin Alzheimer's Disease Research Center.,Geriatric Research Education and Clinical Center, William S. Middleton Memorial VA Hospital, Madison, WI, USA
| | - Elizabeth A Boots
- Wisconsin Alzheimer's Disease Research Center.,Geriatric Research Education and Clinical Center, William S. Middleton Memorial VA Hospital, Madison, WI, USA
| | - Jennifer M Oh
- Wisconsin Alzheimer's Disease Research Center.,Geriatric Research Education and Clinical Center, William S. Middleton Memorial VA Hospital, Madison, WI, USA
| | - Dorothy Edwards
- Department of Kinesiology, University of Wisconsin School of Education, Madison, WI, USA.,Wisconsin Alzheimer's Disease Research Center.,Wisconsin Alzheimer's Institute, Madison, WI, USA
| | - Catherine L Gallagher
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial VA Hospital, Madison, WI, USA.,Wisconsin Alzheimer's Institute, Madison, WI, USA.,Department of Neurology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Cynthia M Carlsson
- Wisconsin Alzheimer's Disease Research Center.,Geriatric Research Education and Clinical Center, William S. Middleton Memorial VA Hospital, Madison, WI, USA.,Wisconsin Alzheimer's Institute, Madison, WI, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center.,Geriatric Research Education and Clinical Center, William S. Middleton Memorial VA Hospital, Madison, WI, USA.,Wisconsin Alzheimer's Institute, Madison, WI, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center.,Geriatric Research Education and Clinical Center, William S. Middleton Memorial VA Hospital, Madison, WI, USA.,Wisconsin Alzheimer's Institute, Madison, WI, USA
| | - Mark A Sager
- Wisconsin Alzheimer's Disease Research Center.,Wisconsin Alzheimer's Institute, Madison, WI, USA
| | - Bruce P Hermann
- Wisconsin Alzheimer's Disease Research Center.,Wisconsin Alzheimer's Institute, Madison, WI, USA
| | - Bradley T Christian
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center.,Geriatric Research Education and Clinical Center, William S. Middleton Memorial VA Hospital, Madison, WI, USA.,Wisconsin Alzheimer's Institute, Madison, WI, USA
| | - Dane B Cook
- Department of Kinesiology, University of Wisconsin School of Education, Madison, WI, USA.,Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Ozioma C Okonkwo
- Wisconsin Alzheimer's Disease Research Center.,Geriatric Research Education and Clinical Center, William S. Middleton Memorial VA Hospital, Madison, WI, USA.,Wisconsin Alzheimer's Institute, Madison, WI, USA
| |
Collapse
|
39
|
Berti V, Walters M, Sterling J, Quinn CG, Logue M, Andrews R, Matthews DC, Osorio RS, Pupi A, Vallabhajosula S, Isaacson RS, de Leon MJ, Mosconi L. Mediterranean diet and 3-year Alzheimer brain biomarker changes in middle-aged adults. Neurology 2018; 90:e1789-e1798. [PMID: 29653991 DOI: 10.1212/wnl.0000000000005527] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 02/26/2018] [Indexed: 01/29/2023] Open
Abstract
OBJECTIVE To examine in a 3-year brain imaging study the effects of higher vs lower adherence to a Mediterranean-style diet (MeDi) on Alzheimer disease (AD) biomarker changes (brain β-amyloid load via 11C-Pittsburgh compound B [PiB] PET and neurodegeneration via 18F-fluorodeoxyglucose [FDG] PET and structural MRI) in midlife. METHODS Seventy 30- to 60-year-old cognitively normal participants with clinical, neuropsychological, and dietary examinations and imaging biomarkers at least 2 years apart were examined. These included 34 participants with higher (MeDi+) and 36 with lower (MeDi-) MeDi adherence. Statistical parametric mapping and volumes of interest were used to compare AD biomarkers between groups at cross section and longitudinally. RESULTS MeDi groups were comparable for clinical and neuropsychological measures. At baseline, compared to the MeDi+ group, the MeDi- group showed reduced FDG-PET glucose metabolism (CMRglc) and higher PiB-PET deposition in AD-affected regions (p < 0.001). Longitudinally, the MeDi--group showed CMRglc declines and PiB increases in these regions, which were greater than those in the MeDi+ group (pinteraction < 0.001). No effects were observed on MRI. Higher MeDi adherence was estimated to provide 1.5 to 3.5 years of protection against AD. CONCLUSION Lower MeDi adherence was associated with progressive AD biomarker abnormalities in middle-aged adults. These data support further investigation of dietary interventions for protection against brain aging and AD.
Collapse
Affiliation(s)
- Valentina Berti
- From the Department of Clinical Pathophysiology (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Department of Nutrition and Food Studies (M.W., L.M.), New York University Steinhardt School of Culture, Education, and Human Development, NY; Woodrow Wilson School of Public and International Affairs (J.S.), Department of Psychology, Princeton University, NJ; Department of Psychiatry (C.G.Q., M.L., R.S.O., L.M.), New York University School of Medicine, NY; ADM Diagnostics (R.A., D.C.M., M.J.d.L.), Chicago, IL; and Departments of Radiology (S.V.) and Neurology (R.S.I., L.M.), Weill Cornell Medical Center/NewYork-Presbyterian, NY
| | - Michelle Walters
- From the Department of Clinical Pathophysiology (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Department of Nutrition and Food Studies (M.W., L.M.), New York University Steinhardt School of Culture, Education, and Human Development, NY; Woodrow Wilson School of Public and International Affairs (J.S.), Department of Psychology, Princeton University, NJ; Department of Psychiatry (C.G.Q., M.L., R.S.O., L.M.), New York University School of Medicine, NY; ADM Diagnostics (R.A., D.C.M., M.J.d.L.), Chicago, IL; and Departments of Radiology (S.V.) and Neurology (R.S.I., L.M.), Weill Cornell Medical Center/NewYork-Presbyterian, NY
| | - Joanna Sterling
- From the Department of Clinical Pathophysiology (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Department of Nutrition and Food Studies (M.W., L.M.), New York University Steinhardt School of Culture, Education, and Human Development, NY; Woodrow Wilson School of Public and International Affairs (J.S.), Department of Psychology, Princeton University, NJ; Department of Psychiatry (C.G.Q., M.L., R.S.O., L.M.), New York University School of Medicine, NY; ADM Diagnostics (R.A., D.C.M., M.J.d.L.), Chicago, IL; and Departments of Radiology (S.V.) and Neurology (R.S.I., L.M.), Weill Cornell Medical Center/NewYork-Presbyterian, NY
| | - Crystal G Quinn
- From the Department of Clinical Pathophysiology (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Department of Nutrition and Food Studies (M.W., L.M.), New York University Steinhardt School of Culture, Education, and Human Development, NY; Woodrow Wilson School of Public and International Affairs (J.S.), Department of Psychology, Princeton University, NJ; Department of Psychiatry (C.G.Q., M.L., R.S.O., L.M.), New York University School of Medicine, NY; ADM Diagnostics (R.A., D.C.M., M.J.d.L.), Chicago, IL; and Departments of Radiology (S.V.) and Neurology (R.S.I., L.M.), Weill Cornell Medical Center/NewYork-Presbyterian, NY
| | - Michelle Logue
- From the Department of Clinical Pathophysiology (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Department of Nutrition and Food Studies (M.W., L.M.), New York University Steinhardt School of Culture, Education, and Human Development, NY; Woodrow Wilson School of Public and International Affairs (J.S.), Department of Psychology, Princeton University, NJ; Department of Psychiatry (C.G.Q., M.L., R.S.O., L.M.), New York University School of Medicine, NY; ADM Diagnostics (R.A., D.C.M., M.J.d.L.), Chicago, IL; and Departments of Radiology (S.V.) and Neurology (R.S.I., L.M.), Weill Cornell Medical Center/NewYork-Presbyterian, NY
| | - Randolph Andrews
- From the Department of Clinical Pathophysiology (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Department of Nutrition and Food Studies (M.W., L.M.), New York University Steinhardt School of Culture, Education, and Human Development, NY; Woodrow Wilson School of Public and International Affairs (J.S.), Department of Psychology, Princeton University, NJ; Department of Psychiatry (C.G.Q., M.L., R.S.O., L.M.), New York University School of Medicine, NY; ADM Diagnostics (R.A., D.C.M., M.J.d.L.), Chicago, IL; and Departments of Radiology (S.V.) and Neurology (R.S.I., L.M.), Weill Cornell Medical Center/NewYork-Presbyterian, NY
| | - Dawn C Matthews
- From the Department of Clinical Pathophysiology (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Department of Nutrition and Food Studies (M.W., L.M.), New York University Steinhardt School of Culture, Education, and Human Development, NY; Woodrow Wilson School of Public and International Affairs (J.S.), Department of Psychology, Princeton University, NJ; Department of Psychiatry (C.G.Q., M.L., R.S.O., L.M.), New York University School of Medicine, NY; ADM Diagnostics (R.A., D.C.M., M.J.d.L.), Chicago, IL; and Departments of Radiology (S.V.) and Neurology (R.S.I., L.M.), Weill Cornell Medical Center/NewYork-Presbyterian, NY
| | - Ricardo S Osorio
- From the Department of Clinical Pathophysiology (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Department of Nutrition and Food Studies (M.W., L.M.), New York University Steinhardt School of Culture, Education, and Human Development, NY; Woodrow Wilson School of Public and International Affairs (J.S.), Department of Psychology, Princeton University, NJ; Department of Psychiatry (C.G.Q., M.L., R.S.O., L.M.), New York University School of Medicine, NY; ADM Diagnostics (R.A., D.C.M., M.J.d.L.), Chicago, IL; and Departments of Radiology (S.V.) and Neurology (R.S.I., L.M.), Weill Cornell Medical Center/NewYork-Presbyterian, NY
| | - Alberto Pupi
- From the Department of Clinical Pathophysiology (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Department of Nutrition and Food Studies (M.W., L.M.), New York University Steinhardt School of Culture, Education, and Human Development, NY; Woodrow Wilson School of Public and International Affairs (J.S.), Department of Psychology, Princeton University, NJ; Department of Psychiatry (C.G.Q., M.L., R.S.O., L.M.), New York University School of Medicine, NY; ADM Diagnostics (R.A., D.C.M., M.J.d.L.), Chicago, IL; and Departments of Radiology (S.V.) and Neurology (R.S.I., L.M.), Weill Cornell Medical Center/NewYork-Presbyterian, NY
| | - Shankar Vallabhajosula
- From the Department of Clinical Pathophysiology (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Department of Nutrition and Food Studies (M.W., L.M.), New York University Steinhardt School of Culture, Education, and Human Development, NY; Woodrow Wilson School of Public and International Affairs (J.S.), Department of Psychology, Princeton University, NJ; Department of Psychiatry (C.G.Q., M.L., R.S.O., L.M.), New York University School of Medicine, NY; ADM Diagnostics (R.A., D.C.M., M.J.d.L.), Chicago, IL; and Departments of Radiology (S.V.) and Neurology (R.S.I., L.M.), Weill Cornell Medical Center/NewYork-Presbyterian, NY
| | - Richard S Isaacson
- From the Department of Clinical Pathophysiology (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Department of Nutrition and Food Studies (M.W., L.M.), New York University Steinhardt School of Culture, Education, and Human Development, NY; Woodrow Wilson School of Public and International Affairs (J.S.), Department of Psychology, Princeton University, NJ; Department of Psychiatry (C.G.Q., M.L., R.S.O., L.M.), New York University School of Medicine, NY; ADM Diagnostics (R.A., D.C.M., M.J.d.L.), Chicago, IL; and Departments of Radiology (S.V.) and Neurology (R.S.I., L.M.), Weill Cornell Medical Center/NewYork-Presbyterian, NY
| | - Mony J de Leon
- From the Department of Clinical Pathophysiology (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Department of Nutrition and Food Studies (M.W., L.M.), New York University Steinhardt School of Culture, Education, and Human Development, NY; Woodrow Wilson School of Public and International Affairs (J.S.), Department of Psychology, Princeton University, NJ; Department of Psychiatry (C.G.Q., M.L., R.S.O., L.M.), New York University School of Medicine, NY; ADM Diagnostics (R.A., D.C.M., M.J.d.L.), Chicago, IL; and Departments of Radiology (S.V.) and Neurology (R.S.I., L.M.), Weill Cornell Medical Center/NewYork-Presbyterian, NY
| | - Lisa Mosconi
- From the Department of Clinical Pathophysiology (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Department of Nutrition and Food Studies (M.W., L.M.), New York University Steinhardt School of Culture, Education, and Human Development, NY; Woodrow Wilson School of Public and International Affairs (J.S.), Department of Psychology, Princeton University, NJ; Department of Psychiatry (C.G.Q., M.L., R.S.O., L.M.), New York University School of Medicine, NY; ADM Diagnostics (R.A., D.C.M., M.J.d.L.), Chicago, IL; and Departments of Radiology (S.V.) and Neurology (R.S.I., L.M.), Weill Cornell Medical Center/NewYork-Presbyterian, NY.
| |
Collapse
|
40
|
Vassilaki M, Aakre JA, Syrjanen JA, Mielke MM, Geda YE, Kremers WK, Machulda MM, Alhurani RE, Staubo SC, Knopman DS, Petersen RC, Lowe VJ, Jack CR, Roberts RO. Mediterranean Diet, Its Components, and Amyloid Imaging Biomarkers. J Alzheimers Dis 2018; 64:281-290. [PMID: 29889074 PMCID: PMC6031931 DOI: 10.3233/jad-171121] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND There is accumulating evidence suggesting that diet may play a role in preventing or delaying cognitive decline and dementia, but the underlying biological mechanisms are not well understood. OBJECTIVES To examine the cross-sectional associations of the Mediterranean diet (MeDi) and its components with 11C-PiB-PET scan measures of amyloid-β (Aβ) deposition. METHODS The study consisted of 278 Mayo Clinic Study of Aging participants 70+ years old, who were cognitively unimpaired (CU) at the time of completion of the Food Frequency Questionnaire (FFQ) and when they underwent PET imaging. Adherence to the MeDi was assessed by computing the MeDi score for each participant. All scans were performed after the FFQ completion; median [IQR] time between FFQ and Aβ PET was 3.5 (1.4) years. Z-scores were created for component, macro- and micronutrients measured. Linear and logistic regression models were adjusted for age, sex, education, apolipoprotein E (APOE) ɛ4 allele carrier status, time interval between the FFQ completion and PET scan, and total energy intake. RESULTS Participants' median age at FFQ was 77.7 years (55.8% men; 26.6% with an APOE ɛ4 allele). Higher MeDi score (linear regression slope (beta):-0.035, p = 0.012; per standard deviation increase), vegetable intake (beta:-0.043, p = 0.002), intake of vitamin A (beta:-0.041, p = 0.003) or β-carotene (beta: -0.039, p = 0.005) from food sources and moderate alcohol consumption (beta: -0.074, p = 0.03) were associated with lower 11C-PiB standardized uptake value ratio. CONCLUSION Findings are consistent with previous studies suggesting that higher adherence to a MeDi pattern and higher vegetable consumption are associated with better neuroimaging biomarker profile. Prospective studies are needed to validate current findings.
Collapse
Affiliation(s)
- Maria Vassilaki
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Jeremiah A Aakre
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Jeremy A Syrjanen
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Michelle M Mielke
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Yonas E Geda
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry and Psychology and Department of Neurology, Mayo Clinic, Scottsdale, AZ, USA
| | - Walter K Kremers
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | | | - Sara C Staubo
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
- Charles University in Prague, Faculty in Hradec Kralove, Hradec Kralove, Czech Republic
| | | | - Ronald C Petersen
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Rosebud O Roberts
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
41
|
Adherence to Mediterranean diet and subjective cognitive function in men. Eur J Epidemiol 2017; 33:223-234. [PMID: 29147948 DOI: 10.1007/s10654-017-0330-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 11/04/2017] [Indexed: 01/04/2023]
Abstract
Benefits of a Mediterranean diet for cognition have been suggested, but epidemiologic studies have been relatively small and of limited duration. To prospectively assess the association between long-term adherence to a Mediterranean dietary pattern and self-reported subjective cognitive function (SCF). Prospective observational study. The Health Professionals' Follow-up Study, a prospective cohort of 51,529 men, 40-75 years of age when enrolled in 1986, of whom 27,842 were included in the primary analysis. Mediterranean diet (MD) score, computed from the mean of five food frequency questionnaires, assessed every 4 years from 1986 to 2002. Self-reported SCF assessed by a 6-item questionnaire in 2008 and 2012, and validated by association with genetic variants in apolipoprotein-4. Using the average of 2008 and 2012 SCF scores, 38.0% of men were considered to have moderate memory scores and 7.3% were considered to have poor scores. In a multivariate model, compared with men having a MD score in the lowest quintile, those in the highest quintile had a 36% lower odds of a poor SCF score (odds ratio 0.64, 95% CI 0.55-0.75; P, trend < 0.001) and a 24% lower odds of a moderate SCF score (OR 0.76, 95% CI 0.70-0.83; P, trend < 0.001). Both remote and more recent diet contributed to this relation. Associations were only slightly weaker using baseline dietary data and a lag of 22 years. Long-term adherence to the Mediterranean diet pattern was strongly related to lower subjective cognitive function. These findings provide further evidence that a healthy dietary pattern may prevent or delay cognitive decline.
Collapse
|
42
|
Zamroziewicz MK, Talukdar MT, Zwilling CE, Barbey AK. Nutritional status, brain network organization, and general intelligence. Neuroimage 2017; 161:241-250. [DOI: 10.1016/j.neuroimage.2017.08.043] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/13/2017] [Accepted: 08/14/2017] [Indexed: 11/25/2022] Open
|
43
|
Anastasiou CA, Yannakoulia M, Kosmidis MH, Dardiotis E, Hadjigeorgiou GM, Sakka P, Arampatzi X, Bougea A, Labropoulos I, Scarmeas N. Mediterranean diet and cognitive health: Initial results from the Hellenic Longitudinal Investigation of Ageing and Diet. PLoS One 2017; 12:e0182048. [PMID: 28763509 PMCID: PMC5538737 DOI: 10.1371/journal.pone.0182048] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 07/11/2017] [Indexed: 12/16/2022] Open
Abstract
Background The Mediterranean dietary pattern has been associated with a decreased risk of many degenerative diseases and cognitive function in particular; however, relevant information from Mediterranean regions, where the prototype Mediterranean diet is typically adhered to, have been very limited. Additionally, predefined Mediterranean diet (MeDi) scores with use of a priori cut-offs have been used very rarely, limiting comparisons between different populations and thus external validity of the associations. Finally, associations between individual components of MeDi (i.e., food groups, macronutrients) and particular aspects of cognitive performance have rarely been explored. We evaluated the association of adherence to an a priori defined Mediterranean dietary pattern and its components with dementia and specific aspects of cognitive function in a representative population cohort in Greece. Methods Participants from the Hellenic Longitudinal Investigation of Ageing and Diet (HELIAD), an on-going population-based study, exploring potential associations between diet and cognitive performance in a representative sample from Greek regions, were included in this analysis. Diagnosis of dementia was made by a full clinical and neuropsychological evaluation, while cognitive performance was assessed according to five cognitive domains (memory, language, attention-speed, executive functioning, visuospatial perception) and a composite cognitive score. Adherence to MeDi was evaluated by an a priori score (range 0–55), derived from a detailed food frequency questionnaire. Results Among 1,865 individuals (mean age 73±6 years, 41% male), 90 were diagnosed with dementia and 223 with mild cognitive impairment. Each unit increase in the Mediterranean dietary score (MedDietScore) was associated with a 10% decrease in the odds for dementia. Adherence to the MeDi was also associated with better performance in memory, language, visuospatial perception and the composite cognitive score; the associations were strongest for memory. Fish consumption was negatively associated with dementia and cognitive performance positively associated with non-refined cereal consumption. Conclusions Our results suggest that adherence to the MeDi is associated with better cognitive performance and lower dementia rates in Greek elders. Thus, the MeDi in its a priori constructed prototype form may have cognitive benefits in traditional Mediterranean populations.
Collapse
Affiliation(s)
- Costas A. Anastasiou
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
- Eginition Hospital, 1st Neurology Clinic, Department of Social Medicine,Psychiatry and Neurology, National and Kapodistrian University of Athens, Athens, Greece
| | - Mary Yannakoulia
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | - Mary H. Kosmidis
- Laboratoty of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | | | - Paraskevi Sakka
- Athens Association of Alzheimer’s Disease and Related Disorders, Marousi, Greece
| | - Xanthi Arampatzi
- Laboratoty of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | | | - Nikolaos Scarmeas
- Eginition Hospital, 1st Neurology Clinic, Department of Social Medicine,Psychiatry and Neurology, National and Kapodistrian University of Athens, Athens, Greece
- Taub Institute for Research in Alzheimer’s Disease and the Aging Brain, the Gertrude H. Sergievsky Center, Department of Neurology, Columbia University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
44
|
|
45
|
Dingova D, Fazekas T, Okuliarova P, Strbova J, Kucera M, Hrabovska A. Low Plasma Cholinesterase Activities are Associated with Deficits in Spatial Orientation, Reduced Ability to Perform Basic Activities of Daily Living, and Low Body Mass Index in Patients with Progressed Alzheimer's Disease. J Alzheimers Dis 2016; 51:801-13. [PMID: 26890780 DOI: 10.3233/jad-151060] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is a progressive and irreversible neurodegenerative disorder characterized by a central cholinergic deficit. Non-neuronal cholinergic changes are, however, described as well. Here we focused on possible changes in the activity of the plasma cholinesterases, acetylcholinesterase (AChE) and butyrylcholinesterase (BChE), in hospitalized AD patients. We analyzed plasma AChE and BChE activities with regards to age, gender, body mass index (BMI), cognitive functions, and ability to perform activities of daily living in AD patients in comparison to healthy subjects. We observed lower AChE activity and trend toward lower BChE activity in AD patients, which both correlated with low BMI. AD patients unable to perform basic activities of daily living (feeding, bathing, dressing, and grooming) showed reduced plasma AChE activities, while worse spatial orientation was linked to lower BChE activities. Three out of four AD patients with the lowest BChE activities died within one year. In conclusion, progressed AD was accompanied by lower plasma AChE activity and trend toward lower BChE activity, which correlated with BMI and deficits in different components of the AD.
Collapse
Affiliation(s)
- Dominika Dingova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia
| | - Tomas Fazekas
- Department of Physical Chemistry of Drugs, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia
| | - Petra Okuliarova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia
| | | | - Matej Kucera
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia
| | - Anna Hrabovska
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia.,Biomedical Research Center, University Hospital, Hradec Kralove, Czech Republic
| |
Collapse
|
46
|
Dao AT, Zagaar MA, Levine AT, Alkadhi KA. Comparison of the Effect of Exercise on Late-Phase LTP of the Dentate Gyrus and CA1 of Alzheimer's Disease Model. Mol Neurobiol 2016; 53:6859-6868. [PMID: 26660327 DOI: 10.1007/s12035-015-9612-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 12/03/2015] [Indexed: 01/15/2023]
Abstract
We investigated the neuroprotective effect of regular treadmill exercise training on long-term memory and its correlate: the late-phase long-term potentiation (L-LTP) and plasticity- and memory-related signaling molecules in the DG and CA1 areas of a rat model of Alzheimer's disease (AD) (i.c.v. infusion of Aβ1-42 peptides, 2 weeks, 250 pmol/day). Testing in the radial arm water maze revealed severe impairment of spatial long-term memory in Aβ-infused sedentary rats but not in exercised Aβ-infused rats. The L-LTP, measured as changes in the field (f)EPSP and in the amplitude of population spike (pspike), was induced by multiple high-frequency stimulation in the CA1 and DG areas of anesthetized rats. The L-LTP of fEPSP in both areas was severely impaired in the sedentary Aβ rats but not in exercised Aβ rats. However, L-LTP of the pspike was severely suppressed in the CA1 area but not in the DG of sedentary Aβ rats. Immunoblot analysis revealed no increase in the levels of phosphorylated (p)-CREB, CaMKIV, and brain-derived neurotrophic factor (BDNF) in both CA1 and DG areas of sedentary Aβ rats during L-LTP, whereas the levels of these molecules were robustly increased in exercised Aβ rats. Impairment of synaptic function may be due to deleterious changes in the molecular signaling cascades that mediate synaptic structural and functional changes. The protective effect of regular exercise can be a promising therapeutic measure for countering or delaying the AD-like pathology.
Collapse
Affiliation(s)
- An T Dao
- Department of PPS, College of Pharmacy, University of Houston, Houston, TX, 77204-5037, USA
| | - Munder A Zagaar
- Department of PPS, College of Pharmacy, University of Houston, Houston, TX, 77204-5037, USA
| | - Amber T Levine
- Department of PPS, College of Pharmacy, University of Houston, Houston, TX, 77204-5037, USA
| | - Karim A Alkadhi
- Department of PPS, College of Pharmacy, University of Houston, Houston, TX, 77204-5037, USA.
| |
Collapse
|
47
|
Steffens EB, Steffens DC. Getting on Message: Preventing Alzheimer Disease Through Diet and Exercise. Am J Geriatr Psychiatry 2016; 24:738-9. [PMID: 27569267 DOI: 10.1016/j.jagp.2016.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 06/02/2016] [Indexed: 10/21/2022]
Affiliation(s)
| | - David C Steffens
- Department of Psychiatry, University of Connecticut Health Center, Farmington, CT
| |
Collapse
|
48
|
Yusufov M, Weyandt LL, Piryatinsky I. Alzheimer's disease and diet: a systematic review. Int J Neurosci 2016; 127:161-175. [DOI: 10.3109/00207454.2016.1155572] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Miryam Yusufov
- Department of Psychology, University of Rhode Island, Kingston, RI, USA
| | - Lisa L. Weyandt
- Department of Psychology, University of Rhode Island, Kingston, RI, USA
| | - Irene Piryatinsky
- Department of Psychiatry and Human Behavior, Butler Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
49
|
Matthews DC, Lukic AS, Andrews RD, Marendic B, Brewer J, Rissman RA, Mosconi L, Strother SC, Wernick MN, Mobley WC, Ness S, Schmidt ME, Rafii MS. Dissociation of Down syndrome and Alzheimer's disease effects with imaging. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2016. [PMID: 28642933 PMCID: PMC5477635 DOI: 10.1016/j.trci.2016.02.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Introduction Down Syndrome (DS) adults experience accumulation of Alzheimer's disease (AD)–like amyloid plaques and tangles and a high incidence of dementia and could provide an enriched population to study AD-targeted treatments. However, to evaluate effects of therapeutic intervention, it is necessary to dissociate the contributions of DS and AD from overall phenotype. Imaging biomarkers offer the potential to characterize and stratify patients who will worsen clinically but have yielded mixed findings in DS subjects. Methods We evaluated 18F fluorodeoxyglucose positron emission tomography (PET), florbetapir PET, and structural magnetic resonance (sMR) image data from 12 nondemented DS adults using advanced multivariate machine learning methods. Results Our results showed distinctive patterns of glucose metabolism and brain volume enabling dissociation of DS and AD effects. AD-like pattern expression corresponded to amyloid burden and clinical measures. Discussion These findings lay groundwork to enable AD clinical trials with characterization and disease-specific tracking of DS adults.
Collapse
Affiliation(s)
| | | | | | | | - James Brewer
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Robert A Rissman
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Lisa Mosconi
- Department of Psychiatry, New York University Langone School of Medicine, New York, NY, USA
| | - Stephen C Strother
- ADM Diagnostics, Northbrook, IL, USA.,Rotman Research Institute, Baycrest Hospital and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Miles N Wernick
- ADM Diagnostics, Northbrook, IL, USA.,Departments of Electrical and Computer Engineering and Biomedical Engineering, Medical Imaging Research Center, Illinois Institute of Technology, Chicago, IL, USA
| | - William C Mobley
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Seth Ness
- Janssen Research and Development LLC, Raritan, NJ, USA
| | | | - Michael S Rafii
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, CA, USA
| |
Collapse
|
50
|
Dubois B, Hampel H, Feldman HH, Scheltens P, Aisen P, Andrieu S, Bakardjian H, Benali H, Bertram L, Blennow K, Broich K, Cavedo E, Crutch S, Dartigues JF, Duyckaerts C, Epelbaum S, Frisoni GB, Gauthier S, Genthon R, Gouw AA, Habert MO, Holtzman DM, Kivipelto M, Lista S, Molinuevo JL, O'Bryant SE, Rabinovici GD, Rowe C, Salloway S, Schneider LS, Sperling R, Teichmann M, Carrillo MC, Cummings J, Jack CR. Preclinical Alzheimer's disease: Definition, natural history, and diagnostic criteria. Alzheimers Dement 2016; 12:292-323. [PMID: 27012484 PMCID: PMC6417794 DOI: 10.1016/j.jalz.2016.02.002] [Citation(s) in RCA: 1276] [Impact Index Per Article: 141.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
During the past decade, a conceptual shift occurred in the field of Alzheimer's disease (AD) considering the disease as a continuum. Thanks to evolving biomarker research and substantial discoveries, it is now possible to identify the disease even at the preclinical stage before the occurrence of the first clinical symptoms. This preclinical stage of AD has become a major research focus as the field postulates that early intervention may offer the best chance of therapeutic success. To date, very little evidence is established on this "silent" stage of the disease. A clarification is needed about the definitions and lexicon, the limits, the natural history, the markers of progression, and the ethical consequence of detecting the disease at this asymptomatic stage. This article is aimed at addressing all the different issues by providing for each of them an updated review of the literature and evidence, with practical recommendations.
Collapse
Affiliation(s)
- Bruno Dubois
- Institute of Memory and Alzheimer's Disease (IM2A) and Brain and Spine Institute (ICM) UMR S 1127 Frontlab, Department of Neurology, AP_HP, Pitié-Salpêtrière University Hospital, Sorbonne Universities, Pierre et Marie Curie University, Paris 06, Paris, France.
| | - Harald Hampel
- Institute of Memory and Alzheimer's Disease (IM2A) and Brain and Spine Institute (ICM) UMR S 1127 Frontlab, Department of Neurology, AP_HP, Pitié-Salpêtrière University Hospital, Sorbonne Universities, Pierre et Marie Curie University, Paris 06, Paris, France; AXA Research Fund & UPMC Chair, Paris, France
| | | | - Philip Scheltens
- Department of Neurology and Alzheimer Center, VU University Medical Center and Neuroscience Campus, Amsterdam, The Netherlands
| | - Paul Aisen
- University of Southern California San Diego, CA, USA
| | - Sandrine Andrieu
- UMR1027, INSERM, Université Toulouse III, Toulouse University Hospital, France
| | - Hovagim Bakardjian
- IHU-A-ICM-Institut des Neurosciences translationnelles de Paris, Paris, France
| | - Habib Benali
- INSERM U1146-CNRS UMR 7371-UPMC UM CR2, Site Pitié-Salpêtrière, Paris, France
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), Institutes of Neurogenetics and Integrative and Experimental Genomics, University of Lübeck, Lübeck, Germany; School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Lab, Department of Neuroscience and Physiology, University of Gothenburg, Mölndal Hospital, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Karl Broich
- Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | - Enrica Cavedo
- AXA Research Fund & UPMC Chair, Paris, France; Laboratory of Alzheimer's Neuroimaging and Epidemiology, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Sebastian Crutch
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | | | - Charles Duyckaerts
- University Pierre et Marie Curie, Assistance Publique des Hôpitaux de Paris, Alzheimer-Prion Team Institut du Cerveau et de la Moelle (ICM), Paris, France
| | - Stéphane Epelbaum
- Institute of Memory and Alzheimer's Disease (IM2A) and Brain and Spine Institute (ICM) UMR S 1127 Frontlab, Department of Neurology, AP_HP, Pitié-Salpêtrière University Hospital, Sorbonne Universities, Pierre et Marie Curie University, Paris 06, Paris, France
| | - Giovanni B Frisoni
- University Hospitals and University of Geneva, Geneva, Switzerland; IRCCS Fatebenefratelli, Brescia, Italy
| | - Serge Gauthier
- McGill Center for Studies in Aging, Douglas Mental Health Research Institute, Montreal, Canada
| | - Remy Genthon
- Fondation pour la Recherche sur Alzheimer, Hôpital Pitié-Salpêtrière, Paris, France
| | - Alida A Gouw
- UMR1027, INSERM, Université Toulouse III, Toulouse University Hospital, France; Department of Clinical Neurophysiology/MEG Center, VU University Medical Center, Amsterdam
| | - Marie-Odile Habert
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, Paris, France; AP-HP, Hôpital Pitié-Salpêtrière, Département de Médecine Nucléaire, Paris, France
| | - David M Holtzman
- Department of Neurology, Washington University, Hope Center for Neurological Disorders, St. Louis, MO, USA; Department of Neurology, Washington University, Knight Alzheimer's Disease Research Center, St. Louis, MO, USA
| | - Miia Kivipelto
- Center for Alzheimer Research, Karolinska Institutet, Department of Geriatric Medicine, Karolinska University Hospital, Stockholm, Sweden; Institute of Clinical Medicine/ Neurology, University of Eastern Finland, Kuopio, Finland
| | | | - José-Luis Molinuevo
- Alzheimer's Disease and Other Cognitive Disorders Unit, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Sid E O'Bryant
- Center for Alzheimer's & Neurodegenerative Disease Research, University of North Texas Health Science Center, TX, USA
| | - Gil D Rabinovici
- Memory & Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Christopher Rowe
- Department of Molecular Imaging, Austin Health, University of Melbourne, Australia
| | - Stephen Salloway
- Memory and Aging Program, Butler Hospital, Alpert Medical School of Brown University, USA; Department of Neurology, Alpert Medical School of Brown University, USA; Department of Psychiatry, Alpert Medical School of Brown University, USA
| | - Lon S Schneider
- Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Reisa Sperling
- Harvard Medical School, Memory Disorders Unit, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Boston, USA; Harvard Medical School, Memory Disorders Unit, Center for Alzheimer Research and Treatment, Massachusetts General Hospital, Boston, USA
| | - Marc Teichmann
- Institute of Memory and Alzheimer's Disease (IM2A) and Brain and Spine Institute (ICM) UMR S 1127 Frontlab, Department of Neurology, AP_HP, Pitié-Salpêtrière University Hospital, Sorbonne Universities, Pierre et Marie Curie University, Paris 06, Paris, France
| | - Maria C Carrillo
- The Alzheimer's Association Division of Medical & Scientific Relations, Chicago, USA
| | - Jeffrey Cummings
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Cliff R Jack
- Department of Radiology, Mayo Clinic, Rochester MN, USA
| |
Collapse
|