1
|
Shang X, Wang W, Tian L, Shi D, Huang Y, Zhang X, Zhu Z, Zhang X, Liu J, Tang S, Hu Y, Ge Z, Yu H, He M. Association of greenspace and natural environment with brain volumes mediated by lifestyle and biomarkers among urban residents. Arch Gerontol Geriatr 2024; 126:105546. [PMID: 38941948 DOI: 10.1016/j.archger.2024.105546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/28/2024] [Accepted: 06/22/2024] [Indexed: 06/30/2024]
Abstract
OBJECTIVES To examine the associaiton between environmental measures and brain volumes and its potential mediators. STUDY DESIGN This was a prospective study. METHODS Our analysis included 34,454 participants (53.4% females) aged 40-73 years at baseline (between 2006 and 2010) from the UK Biobank. Brain volumes were measured using magnetic resonance imaging between 2014 and 2019. RESULTS Greater proximity to greenspace buffered at 1000 m at baseline was associated with larger volumes of total brain measured 8.8 years after baseline assessment (standardized β (95% CI) for each 10% increment in coverage: 0.013(0.005,0.020)), grey matter (0.013(0.006,0.020)), and white matter (0.011(0.004,0.017)) after adjustment for covariates and air pollution. The corresponding numbers for natural environment buffered at 1000 m were 0.010 (0.004,0.017), 0.009 (0.004,0.015), and 0.010 (0.004,0.016), respectively. Similar results were observed for greenspace and natural environment buffered at 300 m. The strongest mediator for the association between greenspace buffered at 1000 m and total brain volume was smoking (percentage (95% CI) of total variance explained: 7.9% (5.5-11.4%)) followed by mean sphered cell volume (3.3% (1.8-5.8%)), vitamin D (2.9% (1.6-5.1%)), and creatinine in blood (2.7% (1.6-4.7%)). Significant mediators combined explained 18.5% (13.2-25.3%) of the association with total brain volume and 32.9% (95% CI: 22.3-45.7%) of the association with grey matter volume. The percentage (95% CI) of the association between natural environment and total brain volume explained by significant mediators combined was 20.6% (14.7-28.1%)). CONCLUSIONS Higher coverage percentage of greenspace and environment may benefit brain health by promoting healthy lifestyle and improving biomarkers including vitamin D and red blood cell indices.
Collapse
Affiliation(s)
- Xianwen Shang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China; Centre for Eye Research Australia, University of Melbourne, Melbourne, VIC 3002, Australia; Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Melbourne, VIC 3050, Australia; School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong, PR China.
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, PR China
| | - Le Tian
- Comprehensive department, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China
| | - Danli Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, PR China; School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong, PR China; Research Centre for SHARP Vision, The Hong Kong Polytechnic University, Kowloon, Hong Kong, PR China
| | - Yu Huang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China
| | - Xueli Zhang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China
| | - Zhuoting Zhu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China; Centre for Eye Research Australia, University of Melbourne, Melbourne, VIC 3002, Australia
| | - Xiayin Zhang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China
| | - Jiahao Liu
- Centre for Eye Research Australia, University of Melbourne, Melbourne, VIC 3002, Australia
| | - Shulin Tang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China
| | - Yijun Hu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China
| | - Zongyuan Ge
- Monash e-Research Center, Faculty of Engineering, Airdoc Research, Nvidia AI Technology Research Center, Monash University, Melbourne, VIC 3800, Australia
| | - Honghua Yu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China.
| | - Mingguang He
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China; Centre for Eye Research Australia, University of Melbourne, Melbourne, VIC 3002, Australia; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, PR China; School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong, PR China; Research Centre for SHARP Vision, The Hong Kong Polytechnic University, Kowloon, Hong Kong, PR China.
| |
Collapse
|
2
|
Beydoun MA, Beydoun HA, Fanelli-Kuczmarski MT, Hu YH, Shaked D, Weiss J, Waldstein SR, Launer LJ, Evans MK, Zonderman AB. Uncovering mediational pathways behind racial and socioeconomic disparities in brain volumes: insights from the UK Biobank study. GeroScience 2024:10.1007/s11357-024-01371-1. [PMID: 39388067 DOI: 10.1007/s11357-024-01371-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/29/2024] [Indexed: 10/15/2024] Open
Abstract
Mediation pathways explaining racial/ethnic and socioeconomic (SES) disparities in structural MRI markers of brain health remain underexplored. We examined racial/ethnic and SES disparities in sMRI markers and tested total, direct, and indirect effects through lifestyle, health-related, and cognition factors using a structural equations modeling approach among 36,184 UK Biobank participants aged 40-70 years at baseline assessment (47% men). Race (non-White vs. White) and lower SES-predicted poorer brain sMRI volumetric outcomes at follow-up, with racial/ethnic disparities in sMRI outcomes involving multiple pathways and SES playing a central role in those pathways. Mediational patterns differed across outcomes, with the SES-sMRI total effect being partially mediated for all outcomes. Over 20% of the total effect (TE) of race/ethnicity on WMH was explained by the indirect effect (IE), by a combination of different pathways going through SES, lifestyle, health-related, and cognition factors. This is in contrast to < 10% for total brain, gray matter (GM), white matter (WM), and frontal GM left/right. Another significant finding is that around 57% of the total effect for SES and the normalized white matter hyperintensity (WMH) was attributed to an indirect effect. This effect encompasses many pathways that involve lifestyle, health-related, and cognitive aspects. Aside from WMH, the percent of TE of SES mediated through various pathways ranged from ~ 5% for WM to > 15% up to 36% for most of the remaining sMRI outcomes, which are composed mainly of GM phenotypes. Race and SES were important determinants of brain volumetric outcomes, with partial mediation of racial/ethnic disparities through SES, lifestyle, health-related, and cognition factors.
Collapse
Affiliation(s)
- May A Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA.
| | - Hind A Beydoun
- Department of Research Programs, Fort Belvoir Community Hospital, Fort Belvoir, VA, 22060, USA
| | - Marie T Fanelli-Kuczmarski
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA
| | - Yi-Han Hu
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA
| | | | - Jordan Weiss
- Stanford Center On Longevity, Stanford University, Stanford, CA, 94305, USA
| | - Shari R Waldstein
- Department of Psychology, University of Maryland Baltimore County, Catonsville, MD, 21250, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA
| |
Collapse
|
3
|
dos Santos JVL, de Melo ISV, Costa CACB, de Almeida LC, Silva DR, Ferro DC, Paula DTC, Macena MDL, Bueno NB. Association Between Ultra-Processed Food Consumption and Cognitive Performance Among Adolescent Students From Underdeveloped Cities in Brazil: A Cross-Sectional Study. Int J Public Health 2024; 69:1607658. [PMID: 39439747 PMCID: PMC11493626 DOI: 10.3389/ijph.2024.1607658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
Objectives The association between ultra-processed foods (UPF) consumption and cognitive performance needs to be better characterized in adolescents, especially in low-income settings, where the cost of human capital is high. This study investigated the association between cognitive performance and UPF in adolescents from the countryside of the Brazilian Northeast. Methods Adolescents (15-18 years old) from three public high schools were included. Food intake was assessed using three 24-hour dietary recalls. The classification of foods as UPF was determined according to the Nova classification. Cognitive performance was evaluated using the Non-Verbal General Intelligence Test. Results 116 adolescents were included, of which 50 (43.1%) showed low cognitive performance. The average energy intake was 1973.5 kcal, with 24.2% coming from UPF. Participants with low cognitive performance consumed 26.5% (95% CI: [22.2; 30.7]%) of daily energy intake from UPF compared to 22.5% ([18.8; 26.2]%) of those with medium-high cognitive performance (P = 0.17), without differences in energy and macronutrient intake. Conclusion Despite similar UPF consumption compared to the Brazilian average, no association was found between UPF consumption and cognitive performance in this low-income adolescent sample.
Collapse
Affiliation(s)
- João Victor Laurindo dos Santos
- Faculty of Nutrition, Federal University of Alagoas, Maceió, Brazil
- Satuba Campus, Federal Institute of Education, Science and Technology of Alagoas, Satuba, Brazil
| | | | - Clara Andrezza Crisóstomo Bezerra Costa
- Satuba Campus, Federal Institute of Education, Science and Technology of Alagoas, Satuba, Brazil
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, Brazil
| | | | | | | | | | | | - Nassib Bezerra Bueno
- Faculty of Nutrition, Federal University of Alagoas, Maceió, Brazil
- Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
4
|
Vaziri Y. The Mediterranean Diet: A powerful defense against Alzheimer disease-A comprehensive review. Clin Nutr ESPEN 2024; 64:160-167. [PMID: 39349103 DOI: 10.1016/j.clnesp.2024.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/22/2024] [Accepted: 09/24/2024] [Indexed: 10/02/2024]
Abstract
Recent studies have explored the impact of lifestyle, particularly diet, on cognitive decline and Alzheimer's disease (AD) risk. The Mediterranean diet has emerged as a potential safeguard, with observational studies indicating it might help defend against cognitive disorders. High adherence is linked with lower cognitive impairment risk, while low adherence elevates the risk for AD. Though these studies suggest connections between the Mediterranean diet and reduced cognitive decline or AD, they do not establish causality. Potential mechanisms might involve vascular factors, glucose/lipid metabolism, and anti-inflammatory effects. Specific Mediterranean diet components like vegetables, fruits, legumes, cereals, fish, and monounsaturated fats might contribute to cognitive benefits. Large-scale randomized controlled trials are needed to ascertain the diet's influence on AD and cognitive health. Currently, the Mediterranean diet cannot be definitively named as a preventive strategy for AD due to insufficient evidence. More research is essential to identify key ingredients and processes that might have preventive effects on AD. In summary, while the Mediterranean diet shows promise against cognitive decline and AD, further research is needed.
Collapse
Affiliation(s)
- Yashar Vaziri
- Department of Nutrition and Dietetics, Sarab Branch, Islamic Azad University, Sarab, Iran.
| |
Collapse
|
5
|
Raeis-Abdollahi E, Raise-Abdullahi P, Rashidy-Pour A, Meamar M, Askari H. Coffee's protective mechanisms against neurodegeneration. PROGRESS IN BRAIN RESEARCH 2024; 288:167-200. [PMID: 39168556 DOI: 10.1016/bs.pbr.2024.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
A widely consumed beverage, coffee has emerged as a potential protective natural agent against neurodegenerative diseases. This chapter explores the intricate mechanisms by which coffee and its bioactive compounds exert neuroprotective effects. The antioxidant properties of coffee polyphenols, such as chlorogenic acid and caffeic acid, mitigate oxidative stress and neuroinflammation. Coffee modulates neurotransmitter systems, including dopaminergic, cholinergic, and glutamatergic pathways implicated in neurodegeneration. Additionally, coffee activates neuroprotective signaling cascades, such as the Nrf2 pathway, and inhibits pro-inflammatory pathways like NF-κB. Coffee components also influence mitochondrial function, biogenesis, and energy metabolism, thereby promoting neuronal survival. Furthermore, coffee suppresses microglial activation and modulates microglial phenotypes, reducing neuroinflammatory responses. Epidemiological studies and clinical trials provide insights into the potential benefits of coffee consumption on cognitive function and neurodegenerative disease risk. However, future research should focus on identifying specific coffee bioactive compounds and their mechanism of action. This chapter highlights the multifaceted neuroprotective mechanisms of coffee, paving the way for future research and potential therapeutic interventions.
Collapse
Affiliation(s)
- Ehsan Raeis-Abdollahi
- Applied Physiology Research Center, Qom Medical Sciences, Islamic Azad University, Qom, Iran; Department of Basic Medical Sciences, Faculty of Medicine, Qom Medical Sciences, Islamic Azad University, Qom, Iran.
| | | | - Ali Rashidy-Pour
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Morvarid Meamar
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Clinical Research Development Unit, Kowsar Educational Research and Therapeutic Hospital, Semnan University of Medical Sciences, Semnan, Iran
| | - Hassan Askari
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
6
|
Contador I, Buch-Vicente B, del Ser T, Llamas-Velasco S, Villarejo-Galende A, Benito-León J, Bermejo-Pareja F. Charting Alzheimer's Disease and Dementia: Epidemiological Insights, Risk Factors and Prevention Pathways. J Clin Med 2024; 13:4100. [PMID: 39064140 PMCID: PMC11278014 DOI: 10.3390/jcm13144100] [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: 05/23/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Alzheimer's disease (AD), the most common cause of dementia, is a complex and multifactorial condition without cure at present. The latest treatments, based on anti-amyloid monoclonal antibodies, have only a modest effect in reducing the progression of cognitive decline in AD, whereas the possibility of preventing AD has become a crucial area of research. In fact, recent studies have observed a decrease in dementia incidence in developed regions such as the US and Europe. However, these trends have not been mirrored in non-Western countries (Japan or China), and the contributing factors of this reduction remain unclear. The Lancet Commission has delineated a constrained classification of 12 risk factors across different life stages. Nevertheless, the scientific literature has pointed to over 200 factors-including sociodemographic, medical, psychological, and sociocultural conditions-related to the development of dementia/AD. This narrative review aims to synthesize the risk/protective factors of dementia/AD. Essentially, we found that risk/protective factors vary between individuals and populations, complicating the creation of a unified prevention strategy. Moreover, dementia/AD explanatory mechanisms involve a diverse array of genetic and environmental factors that interact from the early stages of life. In the future, studies across different population-based cohorts are essential to validate risk/protective factors of dementia. This evidence would help develop public health policies to decrease the incidence of dementia.
Collapse
Affiliation(s)
- Israel Contador
- Department of Basic Psychology, Psychobiology, and Methodology of Behavioral Sciences, Faculty of Psychology, University of Salamanca, 37005 Salamanca, Spain
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, 17117 Stockholm, Sweden
| | - Bárbara Buch-Vicente
- Department of Basic Psychology, Psychobiology, and Methodology of Behavioral Sciences, Faculty of Psychology, University of Salamanca, 37005 Salamanca, Spain
| | - Teodoro del Ser
- Alzheimer Centre Reina Sofia—CIEN Foundation, Institute of Health Carlos III, 28031 Madrid, Spain;
| | - Sara Llamas-Velasco
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.L.-V.); (A.V.-G.); (J.B.-L.)
- Department of Neurology, University Hospital 12 de Octubre, 28041 Madrid, Spain
| | - Alberto Villarejo-Galende
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.L.-V.); (A.V.-G.); (J.B.-L.)
- Department of Neurology, University Hospital 12 de Octubre, 28041 Madrid, Spain
| | - Julián Benito-León
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (S.L.-V.); (A.V.-G.); (J.B.-L.)
- Department of Neurology, University Hospital 12 de Octubre, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
- Department of Medicine, Faculty of Medicine, Complutense University, 28040 Madrid, Spain
| | - Félix Bermejo-Pareja
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
- Department of Medicine, Faculty of Medicine, Complutense University, 28040 Madrid, Spain
| |
Collapse
|
7
|
Soares Dias Portela A, Saxena V, Rosenn E, Wang SH, Masieri S, Palmieri J, Pasinetti GM. Role of Artificial Intelligence in Multinomial Decisions and Preventative Nutrition in Alzheimer's Disease. Mol Nutr Food Res 2024; 68:e2300605. [PMID: 38175857 DOI: 10.1002/mnfr.202300605] [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: 08/23/2023] [Revised: 10/04/2023] [Indexed: 01/06/2024]
Abstract
Alzheimer's disease (AD) affects 50 million people worldwide, an increase of 35 million since 2015, and it is known for memory loss and cognitive decline. Considering the morbidity associated with AD, it is important to explore lifestyle elements influencing the chances of developing AD, with special emphasis on nutritional aspects. This review will first discuss how dietary factors have an impact in AD development and the possible role of Artificial Intelligence (AI) and Machine Learning (ML) in preventative care of AD patients through nutrition. The Mediterranean-DASH diets provide individuals with many nutrient benefits which assists the prevention of neurodegeneration by having neuroprotective roles. Lack of micronutrients, protein-energy, and polyunsaturated fatty acids increase the chance of cognitive decline, loss of memory, and synaptic dysfunction among others. ML software has the ability to design models of algorithms from data introduced to present practical solutions that are accessible and easy to use. It can give predictions for a precise medicine approach to evaluate individuals as a whole. There is no doubt the future of nutritional science lies on customizing diets for individuals to reduce dementia risk factors, maintain overall health and brain function.
Collapse
Affiliation(s)
| | - Vrinda Saxena
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Eric Rosenn
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Shu-Han Wang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Sibilla Masieri
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Joshua Palmieri
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Giulio Maria Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
- Geriatrics Research, Education and Clinical Center, JJ Peters VA Medical Center, Bronx, NY, 10468, USA
| |
Collapse
|
8
|
Yuan Q, Han Y, Fang S, Lei H, Huang H, Lin H, Wu X, Chen R, Chen Z, Chen J, Li H, Liu N, Du H. Sex difference in the association between triglyceride and intracerebral bleeding risk after intravenous thrombolysis for acute ischemic stroke, a multi-center retrospective study. PeerJ 2024; 12:e17558. [PMID: 38938613 PMCID: PMC11210480 DOI: 10.7717/peerj.17558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/21/2024] [Indexed: 06/29/2024] Open
Abstract
Background Whether the relationship of intracerebral bleeding risk with lipid profile may vary by sex remains unclear. This study aims to investigate potential sex differences in the association between lipid profile and the risk of symptomatic intracerebral hemorrhage (sICH) in patients with acute ischemic stroke (AIS) who received intravenous thrombolysis using recombinant tissue plasminogen activator (r-tPA). Methods This multicenter retrospective observational study analyzed patients with AIS treated with intravenous r-tPA. sICH was defined as a worsening of 4 or higher points in the National Institutes of Health Stroke Scale (NIHSS) score within 36 hours after intravenous thrombolysis in any hemorrhage subtype. We assessed the odds ratio (OR) with 95% confidence interval (CI) of lipid profile for sICH for each sex using logistic regression models adjusted for potential confounding factors. Results Of 957 participants (median age 68 (interquartile range, 59-75), men 628 (65.6%)), 56 sICH events (36 (5.7%) in men and 20 (6.1%) in women) were observed. The risk of sICH in men decreased with increasing serum levels of triglyceride after adjustment for confounding factors (vs lowest tertile, medium tertile OR 0.39, 95% CI [0.17-0.91], top tertile OR 0.33, 95% CI [0.13-0.84], overall p = 0.021; per point increase, adjusted OR 0.29, 95% CI [0.13-0.63], p = 0.002). Neither serum levels of total cholesterol nor low-density lipoprotein (LDL) was associated with sICH in men. In women, there was no association between any of the lipid levels and the risk of sICH. Conclusions This study indicated that the association between serum levels of triglyceride and sICH may vary by sex. In men, increased triglyceride levels decrease the risk of sICH; in women, this association was lost. Further studies on the biological mechanisms for sex differences in stroke risk associated with triglyceride are needed.
Collapse
Affiliation(s)
- Qilin Yuan
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Ying Han
- Department of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Shuangfang Fang
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Hanhan Lei
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Huapin Huang
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Huiying Lin
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Xiaomin Wu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Ronghua Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Zhiting Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jie Chen
- Department of Neurology, Fujian Provincial Hospital South Branch, Fuzhou, Fujian Province, China
| | - Hangfeng Li
- Department of Neurology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
| | - Nan Liu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Houwei Du
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
9
|
Thomas A, Ryan CP, Caspi A, Liu Z, Moffitt TE, Sugden K, Zhou J, Belsky DW, Gu Y. Diet, Pace of Biological Aging, and Risk of Dementia in the Framingham Heart Study. Ann Neurol 2024; 95:1069-1079. [PMID: 38407506 PMCID: PMC11102315 DOI: 10.1002/ana.26900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 01/16/2024] [Accepted: 02/05/2024] [Indexed: 02/27/2024]
Abstract
OBJECTIVE People who eat healthier diets are less likely to develop dementia, but the biological mechanism of this protection is not well understood. We tested the hypothesis that healthy diet protects against dementia because it slows the pace of biological aging. METHODS We analyzed Framingham Offspring Cohort data. We included participants ≥60 years-old, free of dementia and having dietary, epigenetic, and follow-up data. We assessed healthy diet as long-term adherence to the Mediterranean-Dash Intervention for Neurodegenerative Delay diet (MIND, over 4 visits spanning 1991-2008). We measured the pace of aging from blood DNA methylation data collected in 2005-2008 using the DunedinPACE epigenetic clock. Incident dementia and mortality were defined using study records compiled from 2005 to 2008 visit through 2018. RESULTS Of n = 1,644 included participants (mean age 69.6, 54% female), n = 140 developed dementia and n = 471 died over 14 years of follow-up. Greater MIND score was associated with slower DunedinPACE and reduced risks for dementia and mortality. Slower DunedinPACE was associated with reduced risks for dementia and mortality. In mediation analysis, slower DunedinPACE accounted for 27% of the diet-dementia association and 57% of the diet-mortality association. INTERPRETATION Findings suggest that slower pace of aging mediates part of the relationship of healthy diet with reduced dementia risk. Monitoring pace of aging may inform dementia prevention. However, a large fraction of the diet-dementia association remains unexplained and may reflect direct connections between diet and brain aging that do not overlap other organ systems. Investigation of brain-specific mechanisms in well-designed mediation studies is warranted. ANN NEUROL 2024;95:1069-1079.
Collapse
Affiliation(s)
- Aline Thomas
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
- Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Calen P. Ryan
- Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| | - Avshalom Caspi
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
| | - Zhonghua Liu
- Department of Biostatistics, Columbia University, New York, NY 10032, USA
| | - Terrie E. Moffitt
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
| | - Karen Sugden
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
| | - Jiayi Zhou
- Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| | - Daniel W. Belsky
- Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY 10032, USA
- Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| | - Yian Gu
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
- Department of Neurology, Columbia University, New York, NY 10032, USA
- Department of Biostatistics, Columbia University, New York, NY 10032, USA
- Gertrude H. Sergievsky Center, Columbia University, New York, NY 10032, USA
| |
Collapse
|
10
|
Nakaki A, Gomez Y, Castro-Barquero S, Conti A, Vellvé K, Casas I, Genero M, Youssef L, Segalés L, Benitez L, Casas R, Vieta E, Bargallo N, Toschi N, Estruch R, Crispi F, Gratacos E, Crovetto F. The Mediterranean Diet in Pregnancy: Implications for Maternal Brain Morphometry in a Secondary Analysis of the IMPACT BCN Randomized Clinical Trial. Nutrients 2024; 16:1604. [PMID: 38892540 PMCID: PMC11174669 DOI: 10.3390/nu16111604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/07/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
INTRODUCTION A Mediterranean diet has positive effects on the brain in mid-older adults; however, there is scarce information on pregnant individuals. We aimed to evaluate the effect of a structured Mediterranean diet intervention on the cortical structure of the maternal brain during pregnancy. METHODS This study was a secondary analysis of the IMPACT BCN, a randomized clinical trial with 1221 high-risk pregnant women randomly allocated into three groups at 19-23 weeks of gestation: Mediterranean diet intervention, a mindfulness-based stress reduction program, or usual care. Maternal brain magnetic resonance imaging was performed during the third trimester of pregnancy in a random subgroup of participants. For this study, data from the Mediterranean diet and usual groups were analyzed. Maternal dietary intake, adherence to the Mediterranean diet and metabolite biomarkers were evaluated using a food frequency questionnaire, a 17-item dietary screener and plasma/urine samples, respectively. RESULTS The cluster-wise analysis showed that the Mediterranean diet group participants (n = 34) had significantly larger surface areas in the right precuneus (90%CI: <0.0001-0.0004, p < 0.001) and left superior parietal (90%CI: 0.026-0.033, p = 0.03) lobules compared to the usual care group participants (n = 37). A larger right precuneus area was associated with high improvements in adherence to the Mediterranean diet, a high intake of walnuts and high concentrations of urinary hydroxytyrosol. A larger left superior parietal area was associated with a high intake of walnuts and high concentrations of urinary hydroxytyrosol. CONCLUSIONS The promotion of a Mediterranean diet during pregnancy has a significant effect on maternal brain structure.
Collapse
Affiliation(s)
- Ayako Nakaki
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08028 Barcelona, Spain
- Institut d’Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Yvan Gomez
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08028 Barcelona, Spain
| | - Sara Castro-Barquero
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08028 Barcelona, Spain
- Department of Internal Medicine Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Institut de Recerca en Nutrició i Seguretat Alimentaria (INSA-UB), University of Barcelona, 08028 Barcelona, Spain
| | - Allegra Conti
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Kilian Vellvé
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08028 Barcelona, Spain
| | - Irene Casas
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08028 Barcelona, Spain
- Institut d’Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Mariona Genero
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08028 Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Lina Youssef
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08028 Barcelona, Spain
- Institut d’Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona Campus, 08036 Barcelona, Spain
| | - Laura Segalés
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08028 Barcelona, Spain
| | - Leticia Benitez
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08028 Barcelona, Spain
- Institut d’Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Rosa Casas
- Department of Internal Medicine Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Institut de Recerca en Nutrició i Seguretat Alimentaria (INSA-UB), University of Barcelona, 08028 Barcelona, Spain
| | - Eduard Vieta
- Department of Psychiatry and Psychology, Hospital Clinic, Neuroscience Institute, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Nuria Bargallo
- Institut d’Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Radiology Department, Center of Image Diagnostic, Hospital Clínic, Facultad de Medicina, Universidad de Barcelona, 08036 Barcelona, Spain
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy
- Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School, Boston, MA 02115, USA
| | - Ramon Estruch
- Department of Internal Medicine Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- Institut de Recerca en Nutrició i Seguretat Alimentaria (INSA-UB), University of Barcelona, 08028 Barcelona, Spain
| | - Fàtima Crispi
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08028 Barcelona, Spain
- Institut d’Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain and Centre for Biomedical Research on Rare Diseases (CIBERER), 08036 Barcelona, Spain
| | - Eduard Gratacos
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08028 Barcelona, Spain
- Institut d’Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain and Centre for Biomedical Research on Rare Diseases (CIBERER), 08036 Barcelona, Spain
| | - Francesca Crovetto
- BCNatal|Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08028 Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin, RD21/0012/0003, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
11
|
Xiao L, Zhou C, Zhang S, Wang Y. A bibliometric analysis on the health behaviors related to mild cognitive impairment. Front Aging Neurosci 2024; 16:1402347. [PMID: 38765772 PMCID: PMC11099246 DOI: 10.3389/fnagi.2024.1402347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 04/19/2024] [Indexed: 05/22/2024] Open
Abstract
Background Mild cognitive impairment (MCI) is commonly defined as a transitional subclinical state between normal aging and dementia. A growing body of research indicates that health behaviors may play a protective role against cognitive decline and could potentially slow down the progression from MCI to dementia. The aim of this study is to conduct a bibliometric analysis of literature focusing on health behaviors and MCI to summarize the factors and evidence regarding the influence of health behaviors on MCI. Methods The study performed a bibliometric analysis by retrieving publications from the Science Citation Index and Social Sciences Citation Index sub-databases within the Web of Science Core Collection. Utilizing VOSviewer and CiteSpace software, a total of 2,843 eligible articles underwent co-citation, co-keywords, and clustering analyses. This methodology aimed to investigate the current status, trends, major research questions, and potential future directions within the research domain. Results The bibliometric analysis indicates that research on healthy behaviors in individuals with MCI originated in 2002 and experienced rapid growth in 2014, reflecting the increasing global interest in this area. The United States emerged as the primary contributor, accounting for more than one-third of the total scientific output with 982 articles. Journals that published the most articles on MCI-related health behaviors included "Journal of Alzheimer's Disease," "Neurobiology of Aging," "Frontiers in Aging Neuroscience," and other geriatrics-related journals. High-impact papers identified by VOSviewer predominantly cover concepts related to MCI, such as diagnostic criteria, assessment, and multifactorial interventions. Co-occurrence keyword analysis highlights five research hotspots in health behavior associated with MCI: exercise, diet, risk factors and preventive measures for dementia, cognitive decline-related biomarkers, and clinical trials. Conclusion This study provides a comprehensive review of literature on health behavior in individuals with MCI, emphasizing influential documents and journals. It outlines research trends and key focal points, offering valuable insights for researchers to comprehend significant contributions and steer future studies.
Collapse
Affiliation(s)
- Liping Xiao
- Department of Information Technology, Jinan University Library, Guangzhou, China
| | - Chunyi Zhou
- School of Nursing, Hubei University of Chinese Medicine, Wuhan, China
| | - Shibo Zhang
- School of Nursing, Hubei University of Chinese Medicine, Wuhan, China
| | - Yuncui Wang
- School of Nursing, Hubei University of Chinese Medicine, Wuhan, China
- Engineering Research Center for Traditional Chinese Medicine Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
| |
Collapse
|
12
|
2024 Alzheimer's disease facts and figures. Alzheimers Dement 2024; 20:3708-3821. [PMID: 38689398 PMCID: PMC11095490 DOI: 10.1002/alz.13809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
This article describes the public health impact of Alzheimer's disease (AD), including prevalence and incidence, mortality and morbidity, use and costs of care and the ramifications of AD for family caregivers, the dementia workforce and society. The Special Report discusses the larger health care system for older adults with cognitive issues, focusing on the role of caregivers and non-physician health care professionals. An estimated 6.9 million Americans age 65 and older are living with Alzheimer's dementia today. This number could grow to 13.8 million by 2060, barring the development of medical breakthroughs to prevent or cure AD. Official AD death certificates recorded 119,399 deaths from AD in 2021. In 2020 and 2021, when COVID-19 entered the ranks of the top ten causes of death, Alzheimer's was the seventh-leading cause of death in the United States. Official counts for more recent years are still being compiled. Alzheimer's remains the fifth-leading cause of death among Americans age 65 and older. Between 2000 and 2021, deaths from stroke, heart disease and HIV decreased, whereas reported deaths from AD increased more than 140%. More than 11 million family members and other unpaid caregivers provided an estimated 18.4 billion hours of care to people with Alzheimer's or other dementias in 2023. These figures reflect a decline in the number of caregivers compared with a decade earlier, as well as an increase in the amount of care provided by each remaining caregiver. Unpaid dementia caregiving was valued at $346.6 billion in 2023. Its costs, however, extend to unpaid caregivers' increased risk for emotional distress and negative mental and physical health outcomes. Members of the paid health care and broader community-based workforce are involved in diagnosing, treating and caring for people with dementia. However, the United States faces growing shortages across different segments of the dementia care workforce due to a combination of factors, including the absolute increase in the number of people living with dementia. Therefore, targeted programs and care delivery models will be needed to attract, better train and effectively deploy health care and community-based workers to provide dementia care. Average per-person Medicare payments for services to beneficiaries age 65 and older with AD or other dementias are almost three times as great as payments for beneficiaries without these conditions, and Medicaid payments are more than 22 times as great. Total payments in 2024 for health care, long-term care and hospice services for people age 65 and older with dementia are estimated to be $360 billion. The Special Report investigates how caregivers of older adults with cognitive issues interact with the health care system and examines the role non-physician health care professionals play in facilitating clinical care and access to community-based services and supports. It includes surveys of caregivers and health care workers, focusing on their experiences, challenges, awareness and perceptions of dementia care navigation.
Collapse
|
13
|
Luo J, Xu X, Sun Y, Lu X, Zhao L. Association of composite dietary antioxidant index with depression and all-cause mortality in middle-aged and elderly population. Sci Rep 2024; 14:9809. [PMID: 38684752 PMCID: PMC11058273 DOI: 10.1038/s41598-024-60322-0] [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: 11/28/2023] [Accepted: 04/22/2024] [Indexed: 05/02/2024] Open
Abstract
Current research has shown an increasing acceptance of interventions for depression through dietary modifications. However, whether composite dietary antioxidant index (CDAI) is associated with depression and all-cause mortality in middle-aged and elderly population remains unknown. This study aimed to explore those associations in American middle-aged and elderly population. Weighted logistic regression models and weighted Cox proportional hazard regression models were used to assess the association of CDAI, covariates, depression, and all-cause mortality, respectively. The stability of the results was also determined by a linear trend test based on CDAI quintiles. Restricted cubic spline curves were employed to test for non-linear relationships. In the model adjusted for all covariates, significant associations were found with the ORs (95% CI) for CDAI and depression [0.77 (0.67, 0.89)] and the HRs (95% CI) for CDAI with all-cause mortality[0.91 (0.83, 1.00)]. Upon conducting restricted cubic spline curves, we found that the association between CDAI and depression was linear, whereas the association between CDAI and all-cause mortality was non-linear with an inflection point of -0.19. Statistical significance was only found before the inflection point. In this study of middle-aged and elderly Americans, CDAI was linearly negatively associated with depression and non-linearly negatively associated with all-cause mortality.
Collapse
Affiliation(s)
- Juanjuan Luo
- University City Hospital, Affiliated Hospital of Shandong University of Chinese Medicine, Jinan, China
| | - Xiying Xu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yiyan Sun
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xixue Lu
- Neck Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, China.
| | - Leiyong Zhao
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
14
|
Ruiz-Rizzo AL, Finke K, Damoiseaux JS, Bartels C, Buerger K, Cosma NC, Dechent P, Dobisch L, Ewers M, Fliessbach K, Frommann I, Glanz W, Goerss D, Hetzer S, Incesoy EI, Janowitz D, Kilimann I, Laske C, van Lent DM, Munk MHJ, Peters O, Priller J, Ramirez A, Rostamzadeh A, Roy N, Scheffler K, Schneider A, Spottke A, Spruth EJ, Teipel S, Wagner M, Wiltfang J, Yakupov R, Jessen F, Duezel E, Perneczky R, Rauchmann BS. Fornix fractional anisotropy mediates the association between Mediterranean diet adherence and memory four years later in older adults without dementia. Neurobiol Aging 2024; 136:99-110. [PMID: 38340637 DOI: 10.1016/j.neurobiolaging.2024.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024]
Abstract
Here, we investigated whether fractional anisotropy (FA) of hippocampus-relevant white-matter tracts mediates the association between baseline Mediterranean diet adherence (MeDiAd) and verbal episodic memory over four years. Participants were healthy older adults with and without subjective cognitive decline and patients with amnestic mild cognitive impairment from the DELCODE cohort study (n = 376; age: 71.47 ± 6.09 years; 48.7 % female). MeDiAd and diffusion data were obtained at baseline. Verbal episodic memory was assessed at baseline and four yearly follow-ups. The associations between baseline MeDiAd and white matter, and verbal episodic memory's mean and rate of change over four years were tested with latent growth curve modeling. Baseline MeDiAd was associated with verbal episodic memory four years later (95 % confidence interval, CI [0.01, 0.32]) but not with its rate of change over this period. Baseline Fornix FA mediated - and, thus, explained - that association (95 % CI [0.002, 0.09]). Fornix FA may be an appropriate response biomarker of Mediterranean diet interventions on verbal memory in older adults.
Collapse
Affiliation(s)
- Adriana L Ruiz-Rizzo
- Department of Neurology, Jena University Hospital, Jena, Germany; Department of Psychology, General and Experimental Psychology Unit, LMU Munich, Munich, Germany.
| | - Kathrin Finke
- Department of Neurology, Jena University Hospital, Jena, Germany; Department of Psychology, General and Experimental Psychology Unit, LMU Munich, Munich, Germany
| | - Jessica S Damoiseaux
- Department of Psychology, Wayne State University, Detroit, MI, USA; Institute of Gerontology, Wayne State University, Detroit, MI, USA
| | - Claudia Bartels
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Goettingen, Germany
| | - Katharina Buerger
- German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Nicoleta Carmen Cosma
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin-Institute of Psychiatry and Psychotherapy, Germany
| | - Peter Dechent
- MR-Research in Neurosciences, Department of Cognitive Neurology, Georg-August-University Goettingen, Germany
| | - Laura Dobisch
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Michael Ewers
- German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Klaus Fliessbach
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; University of Bonn Medical Center, Dept. of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, Bonn, Germany
| | - Ingo Frommann
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; University of Bonn Medical Center, Dept. of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, Bonn, Germany
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Doreen Goerss
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany; Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Stefan Hetzer
- Bernstein Center for Computational Neuroscience, Charité - Universitätsmedizin, Berlin, Germany
| | - Enise I Incesoy
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany; Department for Psychiatry and Psychotherapy, University Clinic Magdeburg, Germany
| | - Daniel Janowitz
- German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany; Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - 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
| | - Debora Melo van Lent
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA; Department of Neurology, Boston University School of Medicine, Boston, MA, USA; The Framingham Heart Study, Framingham, MA, USA
| | - Matthias H J Munk
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany; Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Oliver Peters
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin-Institute of Psychiatry and Psychotherapy, Germany; German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany; Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany; School of Medicine, Technical University of Munich; Department of Psychiatry and Psychotherapy, Munich, Germany; University of Edinburgh and UK DRI, Edinburgh, UK
| | - Alfredo Ramirez
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; University of Bonn Medical Center, Dept. of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, Bonn, Germany; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, San Antonio, TX, USA
| | - Ayda Rostamzadeh
- Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, Germany
| | - Nina Roy
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Klaus Scheffler
- Department for Biomedical Magnetic Resonance, University of Tübingen, Tübingen, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; University of Bonn Medical Center, Dept. of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, Bonn, Germany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; Department of Neurology, University of Bonn, Bonn, Germany
| | - Eike Jakob Spruth
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany; Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany; Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; University of Bonn Medical Center, Dept. of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, Bonn, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Goettingen, Germany; Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Renat Yakupov
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, Germany
| | - Emrah Duezel
- 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; Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - Robert Perneczky
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College, London, UK; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Boris-Stephan Rauchmann
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK; Institute of Neuroradiology, University Hospital, LMU Munich, Germany
| |
Collapse
|
15
|
Wei B, Xu Y, Du Y, Zhou J, Zhong F, Wu C, Lou Y. Feasibility of Using Magnetic Resonance Spectroscopy Test Biomarkers to Diagnose Alzheimer's Disease: Systematic Evaluation and Meta-Analysis. ACTAS ESPANOLAS DE PSIQUIATRIA 2024; 52:161-171. [PMID: 38622011 PMCID: PMC11016455 DOI: 10.62641/aep.v52i2.1552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is the leading cause of dementia, resulting in impairments in memory, cognition, decision-making, and social skills. Thus, accurate preclinical diagnosis of Alzheimer's disease is paramount. The identification of biomarkers for Alzheimer's disease through magnetic resonance spectroscopy (MRS) represents a novel adjunctive diagnostic approach. OBJECTIVE This study conducted a meta-analysis of the diagnostic results of this technology to explore its feasibility and accuracy. METHODS PubMed, Cochrane Library, EMBASE, and Web of Science databases were searched without restrictions, with the search period extending up to July 31, 2022. The search strategy employed a combination of subject headings and keywords. All retrieved documents underwent screening by two researchers, who selected them for meta-analysis. The included literature was analyzed using Review Manager 5.4 software, with corresponding bias maps, forest plots, and summary receiver operating characteristic (SROC) curves generated and analyzed. RESULTS A total of 344 articles were retrieved initially, with 11 articles meeting the criteria for inclusion in the analysis. The analysis encompassed data from approximately 1766 patients. In the forest plot, both sensitivity (95% CI) and specificity (95% CI) approached 1. Examining the true positive rate, false positive rate, true negative rate, and false negative rate, all studies on the summary receiver operating characteristic (SROC) curve clustered in the upper left quadrant, suggesting a very high accuracy of biomarkers detected by MRS for diagnosing Alzheimer's disease. CONCLUSION The detection of biomarkers by MRS demonstrates feasibility and high accuracy in diagnosing AD. This technology holds promise for widespread adoption in the clinical diagnosis of AD in the future.
Collapse
Affiliation(s)
- Bo Wei
- Department of Neurology, Shaoxing People's Hospital, 312000 Shaoxing, Zhejiang, China
| | - Yiqin Xu
- Department of Neurology, Shaoxing People's Hospital, 312000 Shaoxing, Zhejiang, China
| | - Ye Du
- Department of Neurology, Shaoxing People's Hospital, 312000 Shaoxing, Zhejiang, China
| | - Jie Zhou
- Department of Radiology, Shaoxing Seventh People's Hospital (Affiliated Mental Health Center, Medical College of Shaoxing University), 312000 Shaoxing, Zhejiang, China
| | - Fangfang Zhong
- Department of Neurology, Shaoxing People's Hospital, 312000 Shaoxing, Zhejiang, China
| | - Chenglong Wu
- Department of Neurology, Shaoxing People's Hospital, 312000 Shaoxing, Zhejiang, China
| | - Yiping Lou
- Department of Neurology, Shaoxing People's Hospital, 312000 Shaoxing, Zhejiang, China
| |
Collapse
|
16
|
Xiao YL, Gong Y, Qi YJ, Shao ZM, Jiang YZ. Effects of dietary intervention on human diseases: molecular mechanisms and therapeutic potential. Signal Transduct Target Ther 2024; 9:59. [PMID: 38462638 PMCID: PMC10925609 DOI: 10.1038/s41392-024-01771-x] [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: 08/01/2023] [Revised: 02/05/2024] [Accepted: 02/18/2024] [Indexed: 03/12/2024] Open
Abstract
Diet, serving as a vital source of nutrients, exerts a profound influence on human health and disease progression. Recently, dietary interventions have emerged as promising adjunctive treatment strategies not only for cancer but also for neurodegenerative diseases, autoimmune diseases, cardiovascular diseases, and metabolic disorders. These interventions have demonstrated substantial potential in modulating metabolism, disease trajectory, and therapeutic responses. Metabolic reprogramming is a hallmark of malignant progression, and a deeper understanding of this phenomenon in tumors and its effects on immune regulation is a significant challenge that impedes cancer eradication. Dietary intake, as a key environmental factor, can influence tumor metabolism. Emerging evidence indicates that dietary interventions might affect the nutrient availability in tumors, thereby increasing the efficacy of cancer treatments. However, the intricate interplay between dietary interventions and the pathogenesis of cancer and other diseases is complex. Despite encouraging results, the mechanisms underlying diet-based therapeutic strategies remain largely unexplored, often resulting in underutilization in disease management. In this review, we aim to illuminate the potential effects of various dietary interventions, including calorie restriction, fasting-mimicking diet, ketogenic diet, protein restriction diet, high-salt diet, high-fat diet, and high-fiber diet, on cancer and the aforementioned diseases. We explore the multifaceted impacts of these dietary interventions, encompassing their immunomodulatory effects, other biological impacts, and underlying molecular mechanisms. This review offers valuable insights into the potential application of these dietary interventions as adjunctive therapies in disease management.
Collapse
Affiliation(s)
- Yu-Ling Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yue Gong
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ying-Jia Qi
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
17
|
Randolph JJ, Lacritz LH, Colvin MK, Espe-Pfeifer P, Carter KR, Arnett PA, Fox-Fuller J, Aduen PA, Cullum CM, Sperling SA. Integrating Lifestyle Factor Science into Neuropsychological Practice: A National Academy of Neuropsychology Education Paper. Arch Clin Neuropsychol 2024; 39:121-139. [PMID: 37873931 DOI: 10.1093/arclin/acad078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2023] [Indexed: 10/25/2023] Open
Abstract
OBJECTIVE The primary aim of this paper is to review evidence and clinical implications related to lifestyle activities associated with promoting brain and cognitive health. Our review targets four key lifestyle factors: physical activity and exercise, social engagement, cognitively stimulating activity, and consuming Mediterranean-style diets. METHOD We conducted a critical review of the lifestyle factor literature in the four domains listed earlier. We contextualize this literature review by translating findings, when possible, into evidence-based recommendations to consider when providing neuropsychological services. RESULTS There is significant current evidence supporting the role of physical activity and exercise, social engagement, cognitively stimulating activity, and consuming Mediterranean-style diets on positive brain and cognitive health outcomes. While some null findings are present in all four areas reviewed, the weight of the evidence supports the notion that engaging in these activities may promote brain and cognitive functioning. CONCLUSIONS Clinical neuropsychologists can have confidence in recommending engagement in physical activity, social activity, and cognitively stimulating activity, and adhering to a Mediterranean-style diet to promote brain and cognitive health. We discuss limitations in existing lifestyle factor research and future directions to enhance the existing evidence base, including additional research with historically underrepresented groups and individuals with neurological conditions.
Collapse
Affiliation(s)
- John J Randolph
- Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
- Randolph Neuropsychology Associates, PLLC, Lebanon, NH, USA
| | - Laura H Lacritz
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mary K Colvin
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Patricia Espe-Pfeifer
- Department of Psychiatry & Pediatrics, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | | | - Peter A Arnett
- Psychology Department, The Pennsylvania State University, University Park, PA, USA
| | - Joshua Fox-Fuller
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| | - Paula A Aduen
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - C Munro Cullum
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Scott A Sperling
- Center for Neurological Restoration, Department of Neurology, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
18
|
Pellay H, Thomas A, Baillet M, Helmer C, Catheline G, Marmonier C, Samieri C, Féart C. Dairy products and brain structure in French older adults. Br J Nutr 2024; 131:512-520. [PMID: 37694377 PMCID: PMC10784124 DOI: 10.1017/s0007114523001551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 05/17/2023] [Accepted: 06/22/2023] [Indexed: 09/12/2023]
Abstract
Among food groups with putative benefits for brain structures, dairy products (DP) have been poorly studied. The sample included participants without dementia from the ancillary brain imaging study of the Three-City cohort who were aged 65+ years, had their DP intake assessed with a FFQ at baseline and underwent an anatomical scan 3 years (n 343) or 9 years (n 195) after completing the dietary survey. The frequencies of consumption of total DP, milk and cheese were not associated with brain structure. Compared with the lowest frequency, the highest frequency of fresh DP (F-DP) consumption (< 0·5 v. > 1·5 times/d) was significantly associated with a lower medial temporal lobe volume (MTLV) (β = -1·09 cm3, 95 % CI - 1·83, -0·36) 9 years later. In this population-based study of older adults, the consumption of F-DP more than 1·5 times/d was associated with a lower MTLV, which is considered an early biomarker of Alzheimer's disease, 9 years later. This original study should be replicated in different settings before conclusions are drawn.
Collapse
Affiliation(s)
- Hermine Pellay
- Universty of Bordeaux, INSERM, Bordeaux Population Health, UMR1219, F-33000 Bordeaux, France
- CNIEL, Service Recherche Nutrition-Santé, F-75009 Paris, France
| | - Aline Thomas
- Universty of Bordeaux, INSERM, Bordeaux Population Health, UMR1219, F-33000 Bordeaux, France
| | - Marion Baillet
- Universty of Bordeaux, INSERM, Bordeaux Population Health, UMR1219, F-33000 Bordeaux, France
| | - Catherine Helmer
- Universty of Bordeaux, INSERM, Bordeaux Population Health, UMR1219, F-33000 Bordeaux, France
- Clinical and Epidemiological Research Unit, INSERM CIC1401, F-33000 Bordeaux, France
| | - Gwénaëlle Catheline
- Universty of Bordeaux, CNRS, INCIA, UMR5287, F-33000 Bordeaux, France
- Laboratoire Neuroimagerie et vie quotidienne, EPHE-PSL, F-33000 Bordeaux, France
| | | | - Cécilia Samieri
- Universty of Bordeaux, INSERM, Bordeaux Population Health, UMR1219, F-33000 Bordeaux, France
| | - Catherine Féart
- Universty of Bordeaux, INSERM, Bordeaux Population Health, UMR1219, F-33000 Bordeaux, France
| |
Collapse
|
19
|
Xu W, Xu Z, Guo Y, Wu J. Two decades of research on the role of diet in Alzheimer's disease (2003-2023): a bibliometric and visual analysis based on CiteSpace. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2024; 43:9. [PMID: 38233906 PMCID: PMC10795327 DOI: 10.1186/s41043-024-00503-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/10/2024] [Indexed: 01/19/2024]
Abstract
BACKGROUND In recent years, the impact of diet on Alzheimer's disease (AD) as a modifiable lifestyle has attracted widespread attention. We aimed to elucidate the current research status, frontiers, and research trends regarding the role of diet in AD over the past two decades through CiteSpace. METHODS Studies related to AD and diet that were published from January 1, 2003, to June 30, 2023, were retrieved via the Web of Science Core Collection. We imported the study data into CiteSpace for visual analysis of countries, institutions, co-authors, and co-occurring keywords. RESULTS A total of 922 relevant studies were included in our analysis, which found Nikolaos Scarmeas was the most prolific author (13 studies, 1.41%). The results also indicated that USA and Columbia University were the country and institution with the highest number of publications, with 209 (22.67%) and 23 (2.49%), respectively. The keywords that had a burst in the past four years were neuroinflammation, AD, tau, association, and beta. CONCLUSION Talent exchange and regional cooperation are recommended in this study field. The results indicate that the effectiveness of various dietary patterns and mechanisms of dietary interventions using biomarkers and supplementation with refined nutrients will be the main research trends in the future.
Collapse
Affiliation(s)
- Wanyin Xu
- Department of Nutrition, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People's Republic of China
| | - Zhengyanran Xu
- Department of Neurology, Epilepsy Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yi Guo
- Department of Neurology, Epilepsy Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jing Wu
- Department of Nutrition, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People's Republic of China.
| |
Collapse
|
20
|
Reive BS, Lau V, Sánchez-Lafuente CL, Henri-Bhargava A, Kalynchuk LE, Tremblay MÈ, Caruncho HJ. The Inflammation-Induced Dysregulation of Reelin Homeostasis Hypothesis of Alzheimer's Disease. J Alzheimers Dis 2024; 100:1099-1119. [PMID: 38995785 PMCID: PMC11380287 DOI: 10.3233/jad-240088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Alzheimer's disease (AD) accounts for most dementia cases, but we lack a complete understanding of the mechanisms responsible for the core pathology associated with the disease (e.g., amyloid plaque and neurofibrillary tangles). Inflammation has been identified as a key contributor of AD pathology, with recent evidence pointing towards Reelin dysregulation as being associated with inflammation. Here we describe Reelin signaling and outline existing research involving Reelin signaling in AD and inflammation. Research is described pertaining to the inflammatory and immunological functions of Reelin before we propose a mechanism through which inflammation renders Reelin susceptible to dysregulation resulting in the induction and exacerbation of AD pathology. Based on this hypothesis, it is predicted that disorders of both inflammation (including peripheral inflammation and neuroinflammation) and Reelin dysregulation (including disorders associated with upregulated Reelin expression and disorders of Reelin downregulation) have elevated risk of developing AD. We conclude with a description of AD risk in various disorders involving Reelin dysregulation and inflammation.
Collapse
Affiliation(s)
- Brady S Reive
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Victor Lau
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | | | - Alexandre Henri-Bhargava
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Vancouver Island Health Authority, Victoria, BC, Canada
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Lisa E Kalynchuk
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Mental Health Research Cluster, University of Victoria, Victoria, BC, Canada
| | - Hector J Caruncho
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Mental Health Research Cluster, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
21
|
Khaled M, Al-Jamal H, Tajer L, El-Mir R. Alzheimer's Disease in Lebanon: Exploring Genetic and Environmental Risk Factors-A Comprehensive Review. J Alzheimers Dis 2024; 99:21-40. [PMID: 38640157 DOI: 10.3233/jad-231432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition that displays a high prevalence in Lebanon causing a local burden in healthcare and socio-economic sectors. Unfortunately, the lack of prevalence studies and clinical trials in Lebanon minimizes the improvement of AD patient health status. In this review, we include over 155 articles to cover the different aspects of AD ranging from mechanisms to possible treatment and management tools. We highlight some important modifiable and non-modifiable risk factors of the disease including genetics, age, cardiovascular diseases, smoking, etc. Finally, we propose a hypothetical genetic synergy model between APOE4 and TREM2 genes which constitutes a potential early diagnostic tool that helps in reducing the risk of AD based on preventative measures decades before cognitive decline. The studies on AD in Lebanon and the Middle East are scarce. This review points out the importance of genetic mapping in the understanding of disease pathology which is crucial for the emergence of novel diagnostic tools. Hence, we establish a rigid basis for further research to identify the most influential genetic and environmental risk factors for the purpose of using more specific diagnostic tools and possibly adopting a local management protocol.
Collapse
Affiliation(s)
| | - Hadi Al-Jamal
- Faculty of Public Health III, Lebanese University, Tripoli, Lebanon
| | - Layla Tajer
- Faculty of Public Health III, Lebanese University, Tripoli, Lebanon
| | - Reem El-Mir
- Faculty of Public Health III, Lebanese University, Tripoli, Lebanon
| |
Collapse
|
22
|
Chen M, Wang F, Lei H, Yang Z, Li C. In Silico Insights into Micro-Mechanism Understanding of Extracts of Taxus Chinensis Fruits Against Alzheimer's Disease. J Alzheimers Dis 2024; 97:727-740. [PMID: 38217605 DOI: 10.3233/jad-231066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2024]
Abstract
BACKGROUND The taxus chinensis fruit (TCF) shows promises in treatment of aging-related diseases such as Alzheimer's disease (AD). However, its related constituents and targets against AD have not been deciphered. OBJECTIVE This study was to uncover constituents and targets of TCF extracts against AD. METHODS An integrated approach including ultrasound extractions and constituent identification of TCF by UPLC-QE-MS/MS, target identification of constituents and AD by R data-mining from Pubchem, Drugbank and GEO databases, network construction, molecular docking and the ROC curve analysis was carried out. RESULTS We identified 250 compounds in TCF extracts, and obtained 3,231 known constituent targets and 5,326 differential expression genes of AD, and 988 intersection genes. Through the network construction and KEGG pathway analysis, 19 chemicals, 31 targets, and 11 biological pathways were obtained as core compounds, targets and pathways of TCF extracts against AD. Among these constituents, luteolin, oleic acid, gallic acid, baicalein, naringenin, lovastatin and rutin had obvious anti-AD effect. Molecular docking results further confirmed above results. The ROC AUC values of about 87% of these core targets of TCF extracts was greater than 0.5 in the two GEO chips of AD, especially 10 targets with ROC AUC values greater than 0.7, such as BCL2, CASP7, NFKBIA, HMOX1, CDK2, LDLR, RELA, and CCL2, which mainly referred to neuron apoptosis, response to oxidative stress and inflammation, fibroblast proliferation, etc.Conclusions:The TCF extracts have diverse active compounds that can act on the diagnostic genes of AD, which deserve further in-depth study.
Collapse
Affiliation(s)
- Meimei Chen
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of TCM Health Status Identification, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Fengzhen Wang
- Certification Center for Chinese Physicians, State Administration of Traditional Chinese Medicine, Beijing, Beijing, China
| | - Huangwei Lei
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of TCM Health Status Identification, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Zhaoyang Yang
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of TCM Health Status Identification, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Candong Li
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of TCM Health Status Identification, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| |
Collapse
|
23
|
De Marchi F, Vignaroli F, Mazzini L, Comi C, Tondo G. New Insights into the Relationship between Nutrition and Neuroinflammation in Alzheimer's Disease: Preventive and Therapeutic Perspectives. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:614-627. [PMID: 37291780 DOI: 10.2174/1871527322666230608110201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 04/16/2023] [Accepted: 05/08/2023] [Indexed: 06/10/2023]
Abstract
Neurodegenerative diseases are progressive brain disorders characterized by inexorable synaptic dysfunction and neuronal loss. Since the most consistent risk factor for developing neurodegenerative diseases is aging, the prevalence of these disorders is intended to increase with increasing life expectancy. Alzheimer's disease is the most common cause of neurodegenerative dementia, representing a significant medical, social, and economic burden worldwide. Despite growing research to reach an early diagnosis and optimal patient management, no disease-modifying therapies are currently available. Chronic neuroinflammation has been recognized as a crucial player in sustaining neurodegenerative processes, along with pathological deposition of misfolded proteins, including amyloid-β and tau protein. Modulating neuroinflammatory responses may be a promising therapeutic strategy in future clinical trials. Among factors that are able to regulate neuroinflammatory mechanisms, diet, and nutrients represent easily accessible and modifiable lifestyle components. Mediterranean diet and several nutrients, including polyphenols, vitamins, and omega-3 polyunsaturated fatty acids, can exert antioxidant and anti-inflammatory properties, impacting clinical manifestations, cognitive decline, and dementia. This review aims to provide an updated overview of the relationship between neuroinflammation, nutrition, gut microbiota, and neurodegeneration. We summarize the major studies exploring the effects of diet regimes on cognitive decline, primarily focusing on Alzheimer's disease dementia and the impact of these results on the design of ongoing clinical trials.
Collapse
Affiliation(s)
- Fabiola De Marchi
- ALS Center, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100, Novara, Italy
| | - Francesca Vignaroli
- Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100, Novara, Italy
| | - Letizia Mazzini
- ALS Center, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100, Novara, Italy
| | - Cristoforo Comi
- Neurology Unit, Department of Translational Medicine, S. Andrea Hospital, University of Piemonte Orientale, 13100, Vercelli, Italy
| | - Giacomo Tondo
- Neurology Unit, Department of Translational Medicine, S. Andrea Hospital, University of Piemonte Orientale, 13100, Vercelli, Italy
| |
Collapse
|
24
|
Huang J, Yu Y, Li H, Wei Y, Sun M. Effect of dietary protein intake on cognitive function in the elderly with chronic kidney disease: analysis of the National Health and Nutrition Examination Survey 2011-2014. Ren Fail 2023; 45:2294147. [PMID: 38097960 PMCID: PMC10732213 DOI: 10.1080/0886022x.2023.2294147] [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: 08/22/2023] [Accepted: 12/07/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Cognitive dysfunction is prevalent among the elderly diagnosed with chronic kidney disease (CKD). Low protein diets are used for retarding the progression of CKD in clinical practice. Nonetheless, the impact of dietary protein consumption on cognitive function in this population remains uncertain. METHODS We recruited 2306 participants (≥60 years) from 2011 to 2014 National Health and Nutrition Examination Survey (NHANES). 24-h dietary recall questionnaire was utilized to evaluate protein intake. Cognitive function was measured using the Consortium to Establish a Registry for Alzheimer's Disease (CERAD), Animal Fluency Test (AFT), and Digit Symbol Substitution Test (DSST). Participants' characteristics were analyzed, and the interaction between protein consumption and CKD on cognitive impairment were analyzed using a logistic regression model. RESULTS We divided participants into three groups based on CKD stages: no CKD, CKD stage G1 to G2 (19%), and CKD stage G3 to G5 (18%). The average protein intake was 0.97 g/(kg·d). In the higher protein intake group, CKD stages G1 to G2 elevated the risk of immediate memory impairment (OR: 2.441, 95% Cl: 1.161-5.132 for protein consumption in 1.0-1.2g/(kg·d); OR: 2.225, 95% Cl: 1.015-4.876 for protein consumption in >1.2 g/(kg·d)). However, no similar resuts were observed in the lower protein intake group. In addition, the interaction between CKD status and protein intake on immediate memory was statistically significant (p = .041). CONCLUSION A higher probability of cognitive impairment in the elderly with early-stage CKD may be linked to higher protein intake. Low protein diets may be a potential strategy to release cognitive impairment in the elderly with early-stage CKD.
Collapse
Affiliation(s)
- Jingda Huang
- Department of Nephrology, First Hospital of Jilin University, Changchun, China
| | - Yang Yu
- The Fourth Clinical Medical College, Hebei Medical University, Shijiazhuang, China
| | - Huimin Li
- Department of Nephrology, First Hospital of Jilin University, Changchun, China
| | - Yihui Wei
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Mindan Sun
- Department of Nephrology, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
25
|
Liu J, van Beusekom H, Bu X, Chen G, Henrique Rosado de Castro P, Chen X, Chen X, Clarkson AN, Farr TD, Fu Y, Jia J, Jolkkonen J, Kim WS, Korhonen P, Li S, Liang Y, Liu G, Liu G, Liu Y, Malm T, Mao X, Oliveira JM, Modo MM, Ramos‐Cabrer P, Ruscher K, Song W, Wang J, Wang X, Wang Y, Wu H, Xiong L, Yang Y, Ye K, Yu J, Zhou X, Zille M, Masters CL, Walczak P, Boltze J, Ji X, Wang Y. Preserving cognitive function in patients with Alzheimer's disease: The Alzheimer's disease neuroprotection research initiative (ADNRI). NEUROPROTECTION 2023; 1:84-98. [PMID: 38223913 PMCID: PMC10783281 DOI: 10.1002/nep3.23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 01/16/2024]
Abstract
The global trend toward aging populations has resulted in an increase in the occurrence of Alzheimer's disease (AD) and associated socioeconomic burdens. Abnormal metabolism of amyloid-β (Aβ) has been proposed as a significant pathomechanism in AD, supported by results of recent clinical trials using anti-Aβ antibodies. Nonetheless, the cognitive benefits of the current treatments are limited. The etiology of AD is multifactorial, encompassing Aβ and tau accumulation, neuroinflammation, demyelination, vascular dysfunction, and comorbidities, which collectively lead to widespread neurodegeneration in the brain and cognitive impairment. Hence, solely removing Aβ from the brain may be insufficient to combat neurodegeneration and preserve cognition. To attain effective treatment for AD, it is necessary to (1) conduct extensive research on various mechanisms that cause neurodegeneration, including advances in neuroimaging techniques for earlier detection and a more precise characterization of molecular events at scales ranging from cellular to the full system level; (2) identify neuroprotective intervention targets against different neurodegeneration mechanisms; and (3) discover novel and optimal combinations of neuroprotective intervention strategies to maintain cognitive function in AD patients. The Alzheimer's Disease Neuroprotection Research Initiative's objective is to facilitate coordinated, multidisciplinary efforts to develop systemic neuroprotective strategies to combat AD. The aim is to achieve mitigation of the full spectrum of pathological processes underlying AD, with the goal of halting or even reversing cognitive decline.
Collapse
Affiliation(s)
- Jie Liu
- Department of Neurology, Daping HospitalThird Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Heleen van Beusekom
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MCUniversity Medical CenterRotterdamThe Netherlands
| | - Xian‐Le Bu
- Department of Neurology, Daping HospitalThird Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
| | - Gong Chen
- Guangdong‐HongKong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouGuangdongChina
| | | | - Xiaochun Chen
- Fujian Key Laboratory of Molecular Neurology, Department of Neurology and Geriatrics, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Institute of NeuroscienceFujian Medical UniversityFuzhouFujianChina
| | - Xiaowei Chen
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
- Guangyang Bay LaboratoryChongqing Institute for Brain and IntelligenceChongqingChina
- Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghaiChina
| | - Andrew N. Clarkson
- Department of Anatomy, Brain Health Research Centre and Brain Research New ZealandUniversity of OtagoDunedinNew Zealand
| | - Tracy D. Farr
- School of Life SciencesUniversity of NottinghamNottinghamUK
| | - Yuhong Fu
- Brain and Mind Centre & School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, National Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingChina
| | - Jukka Jolkkonen
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Woojin Scott Kim
- Brain and Mind Centre & School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Paula Korhonen
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan HospitalCapital Medical UniversityBeijingChina
| | - Yajie Liang
- Department of Diagnostic Radiology and Nuclear MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Guang‐Hui Liu
- University of Chinese Academy of SciencesBeijingChina
- State Key Laboratory of Membrane Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Guiyou Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Yu‐Hui Liu
- Department of Neurology, Daping HospitalThird Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Xiaobo Mao
- Institute for Cell Engineering, Department of NeurologyThe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Joaquim Miguel Oliveira
- 3B's Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineUniversity of MinhoGuimarãesPortugal
- ICVS/3B's—PT Government Associate LaboratoryBraga/GuimarãesPortugal
| | - Mike M. Modo
- Department of Bioengineering, McGowan Institute for Regenerative MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Radiology, McGowan Institute for Regenerative MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Pedro Ramos‐Cabrer
- Magnetic Resonance Imaging LaboratoryCIC BiomaGUNE Research Center, Basque Research and Technology Alliance (BRTA)Donostia‐San SebastianSpain
| | - Karsten Ruscher
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical SciencesLund UniversityLundSweden
| | - Weihong Song
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province. Zhejiang Clinical Research Center for Mental Disorders, School of Mental Health and The Affiliated Kangning Hospital, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou Medical UniversityZhejiangChina
| | - Jun Wang
- Department of Neurology, Daping HospitalThird Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Xuanyue Wang
- School of Optometry and Vision ScienceUniversity of New South WalesSydneyNew South WalesAustralia
| | - Yun Wang
- Neuroscience Research Institute, Department of Neurobiology, School of Basic, Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National, Health Commission and State Key Laboratory of Natural and Biomimetic DrugsPeking UniversityBeijingChina
- PKU‐IDG/McGovern Institute for Brain ResearchPeking UniversityBeijingChina
| | - Haitao Wu
- Department of NeurobiologyBeijing Institute of Basic Medical SciencesBeijingChina
| | - Lize Xiong
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain‐Like Intelligence, Shanghai Fourth People's Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Yi Yang
- Department of NeurologyThe First Hospital of Jilin University, Chang ChunJilinChina
| | - Keqiang Ye
- Faculty of Life and Health SciencesBrain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced TechnologyShenzhenChina
| | - Jin‐Tai Yu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xin‐Fu Zhou
- Division of Health Sciences, School of Pharmacy and Medical Sciences and Sansom InstituteUniversity of South AustraliaAdelaideSouth AustraliaAustralia
- Suzhou Auzone BiotechSuzhouJiangsuChina
| | - Marietta Zille
- Department of Pharmaceutical Sciences, Division of Pharmacology and ToxicologyUniversity of ViennaViennaAustria
| | - Colin L. Masters
- The Florey InstituteThe University of Melbourne, ParkvilleVictoriaAustralia
| | - Piotr Walczak
- Department of Diagnostic Radiology and Nuclear MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | | | - Xunming Ji
- Department of NeurosurgeryXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Yan‐Jiang Wang
- Department of Neurology, Daping HospitalThird Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
- Guangyang Bay LaboratoryChongqing Institute for Brain and IntelligenceChongqingChina
- Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghaiChina
| |
Collapse
|
26
|
Gregory S, Blennow K, Ritchie CW, Shannon OM, Stevenson EJ, Muniz-Terrera G. Mediterranean diet is associated with lower white matter lesion volume in Mediterranean cities and lower cerebrospinal fluid Aβ 42 in non-Mediterranean cities in the EPAD LCS cohort. Neurobiol Aging 2023; 131:29-38. [PMID: 37572525 DOI: 10.1016/j.neurobiolaging.2023.07.012] [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: 02/09/2023] [Revised: 07/12/2023] [Accepted: 07/15/2023] [Indexed: 08/14/2023]
Abstract
The Mediterranean diet (MedDiet) has been associated with better brain health and reduced incidence of dementia. Few studies have compared the effects of the MedDiet in early Alzheimer's disease or compared the effects of the diet within and outside of the Mediterranean region. The Mediterranean diet adherence screener (MEDAS) and MEDAS continuous scores were calculated at the baseline visit of the European Prevention of Alzheimer's Dementia Longitudinal Cohort Study (n = 1625). The scores were included in linear regression models to test for associations with hippocampal volume, log-transformed white matter lesion volume, cerebrospinal fluid pTau18, and Aβ42. Higher MEDAS scores were associated with lower log-transformed white matter lesion volume (β: -0.07, standard error [SE]: 0.02, p < 0.001). This association was only seen in the Mediterranean region (β: -0.12, SE: 0.03, p < 0.001). In the non-Mediterranean region, higher MEDAS continuous scores were associated with lower cerebrospinal fluid Aβ42 (β: -68.30, SE: 14.32, p < 0.001). More research is needed to understand the differences in the associations seen with the MedDiet and Alzheimer's disease biomarkers in different European regions.
Collapse
Affiliation(s)
- Sarah Gregory
- Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Craig W Ritchie
- Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; Scottish Brain Sciences, Edinburgh, UK
| | - Oliver M Shannon
- Faculty of Medical Sciences, Human Nutrition Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Emma J Stevenson
- Faculty of Medical Sciences, Human Nutrition Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Graciela Muniz-Terrera
- Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; Department of Social Medicine, Ohio University, Athens, OH, USA
| |
Collapse
|
27
|
Jiang J, Shi H, Jiang S, Wang A, Zou X, Wang Y, Li W, Zhang Y, Sun M, Ren Q, Xu J. Nutrition in Alzheimer's disease: a review of an underappreciated pathophysiological mechanism. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2257-2279. [PMID: 37058185 DOI: 10.1007/s11427-022-2276-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/16/2023] [Indexed: 04/15/2023]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in older individuals and is an escalating challenge to global public health. Pharmacy therapy of AD is one of the well-funded areas; however, little progress has been made due to the complex pathogenesis. Recent evidence has demonstrated that modifying risk factors and lifestyle may prevent or delay the incidence of AD by 40%, which suggests that the management should pivot from single pharmacotherapy toward a multipronged approach because AD is a complex and multifaceted disease. Recently, the gut-microbiota-brain axis has gained tremendous traction in the pathogenesis of AD through bidirectional communication with multiple neural, immune, and metabolic pathways, providing new insights into novel therapeutic strategies. Dietary nutrition is an important and profound environmental factor that influences the composition and function of the microbiota. The Nutrition for Dementia Prevention Working Group recently found that dietary nutrition can affect cognition in AD-related dementia directly or indirectly through complex interactions of behavioral, genetic, systemic, and brain factors. Thus, considering the multiple etiologies of AD, nutrition represents a multidimensional factor that has a profound effect on AD onset and development. However, mechanistically, the effect of nutrition on AD is uncertain; therefore, optimal strategies or the timing of nutritional intervention to prevent or treat AD has not been established.Thus, this review summarizes the current state of knowledge concerning nutritional disorders, AD patient and caregiver burden, and the roles of nutrition in the pathophysiology of AD. We aim to emphasize knowledge gaps to provide direction for future research and to establish optimal nutrition-based intervention strategies for AD.
Collapse
Affiliation(s)
- Jiwei Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Hanping Shi
- Department of Gastrointestinal Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China
| | - Shirui Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Anxin Wang
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Xinying Zou
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Yanli Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Wenyi Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Yuan Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Mengfan Sun
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Qiwei Ren
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Jun Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China.
| |
Collapse
|
28
|
Dhillon VS, Thomas P, Lee SL, Deo P, Fenech M. Red Blood Cell Fatty Acid Profiles Are Significantly Altered in South Australian Mild Cognitive Impairment and Alzheimer's Disease Cases Compared to Matched Controls. Int J Mol Sci 2023; 24:14164. [PMID: 37762467 PMCID: PMC10531649 DOI: 10.3390/ijms241814164] [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: 07/28/2023] [Revised: 09/10/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Nutritional imbalances have been associated with a higher risk for cognitive impairment. This study determined the red blood cell (RBC) fatty acid profile of newly diagnosed mild cognitive impairment (MCI) and Alzheimer's disease (AD) patients compared to age and gender-matched controls. There was a significant increase in palmitic acid (p < 0.00001) for both MCI and AD groups. Saturated fatty acids were significantly elevated in the MCI group, including stearic acid (p = 0.0001), arachidic acid (p = 0.003), behenic acid (p = 0.0002), tricosanoic acid (p = 0.007) and lignoceric acid (p = 0.001). n-6 polyunsaturated fatty acids (PUFAs) were significantly reduced in MCI, including linoleic acid (p = 0.001), γ-linolenic acid (p = 0.03), eicosatrienoic acid (p = 0.009) and arachidonic acid (p < 0.00004). The n-3 PUFAs, α-linolenic acid and docosahexaenoic acid, were both significantly reduced in MCI and AD (p = 0.0005 and p = 0.00003). A positive correlation was evident between the Mini-Mental State Examination score and nervonic acid in MCI (r = 0.54, p = 0.01) and a negative correlation with γ-linolenic acid in AD (r = -0.43, p = 0.05). Differences in fatty acid profiles may prove useful as potential biomarkers reflecting increased risk for dementia.
Collapse
Affiliation(s)
- Varinderpal S. Dhillon
- Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia; (P.D.); (M.F.)
| | - Philip Thomas
- CSIRO Health and Biosecurity, Adelaide 5000, Australia;
| | - Sau L. Lee
- College of Medical and Public Health, Flinders University, Bedford Park 5042, Australia;
| | - Permal Deo
- Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia; (P.D.); (M.F.)
| | - Michael Fenech
- Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia; (P.D.); (M.F.)
- Genome Health Foundation, Adelaide 5048, Australia
| |
Collapse
|
29
|
Zhou Y, Xie L, Schröder J, Schuster IS, Nakai M, Sun G, Sun YBY, Mariño E, Degli-Esposti MA, Marques FZ, Grubman A, Polo JM, Mackay CR. Dietary Fiber and Microbiota Metabolite Receptors Enhance Cognition and Alleviate Disease in the 5xFAD Mouse Model of Alzheimer's Disease. J Neurosci 2023; 43:6460-6475. [PMID: 37596052 PMCID: PMC10506626 DOI: 10.1523/jneurosci.0724-23.2023] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/20/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with poorly understood etiology. AD has several similarities with other "Western lifestyle" inflammatory diseases, where the gut microbiome and immune pathways have been associated. Previously, we and others have noted the involvement of metabolite-sensing GPCRs and their ligands, short-chain fatty acids (SCFAs), in protection of numerous Western diseases in mouse models, such as Type I diabetes and hypertension. Depletion of GPR43, GPR41, or GPR109A accelerates disease, whereas high SCFA yielding diets protect in mouse models. Here, we extended the concept that metabolite-sensing receptors and SCFAs may be a more common protective mechanism against Western diseases by studying their role in AD pathogenesis in the 5xFAD mouse model. Both male and female mice were included. Depletion of GPR41 and GPR43 accelerated cognitive decline and impaired adult hippocampal neurogenesis in 5xFAD and WT mice. Lack of fiber/SCFAs accelerated a memory deficit, whereas diets supplemented with high acetate and butyrate (HAMSAB) delayed cognitive decline in 5xFAD mice. Fiber intake impacted on microglial morphology in WT mice and microglial clustering phenotype in 5xFAD mice. Lack of fiber impaired adult hippocampal neurogenesis in both W and AD mice. Finally, maternal dietary fiber intake significantly affects offspring's cognitive functions in 5xFAD mice and microglial transcriptome in both WT and 5xFAD mice, suggesting that SCFAs may exert their effect during pregnancy and lactation. Together, metabolite-sensing GPCRs and SCFAs are essential for protection against AD, and reveal a new strategy for disease prevention.Significance Statement Alzheimer's disease (AD) is one of the most common neurodegenerative diseases; currently, there is no cure for AD. In our study, short-chain fatty acids and metabolite receptors play an important role in cognitive function and pathology in AD mouse model as well as in WT mice. SCFAs also impact on microglia transcriptome, and immune cell recruitment. Out study indicates the potential of specialized diets (supplemented with high acetate and butyrate) releasing high amounts of SCFAs to protect against disease.
Collapse
Affiliation(s)
- Yichen Zhou
- Department of Microbiology, Monash University, Clayton, Victoria, Australia, 3800
| | - Liang Xie
- Department of Microbiology, Monash University, Clayton, Victoria, Australia, 3800
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Clayton, Victoria, Australia, 3800
| | - Jan Schröder
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia, 3800
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia, 3800
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia, 3800
| | - Iona S Schuster
- Department of Microbiology, Monash University, Clayton, Victoria, Australia, 3800
- Center for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia, 6009
| | - Michael Nakai
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Clayton, Victoria, Australia, 3800
| | - Guizhi Sun
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia, 3800
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia, 3800
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia, 3800
| | - Yu B Y Sun
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia, 3800
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia, 3800
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia, 3800
| | - Eliana Mariño
- Department of Biochemistry & Molecular Biology, Monash University, Clayton, Victoria, Australia, 3800
| | - Mariapia A Degli-Esposti
- Department of Microbiology, Monash University, Clayton, Victoria, Australia, 3800
- Center for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia, 6009
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Clayton, Victoria, Australia, 3800
- Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia, 6009
| | - Alexandra Grubman
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia, 3800
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia, 3800
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia, 3800
| | - Jose M Polo
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia, 3800
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia, 3800
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia, 3800
| | - Charles R Mackay
- Department of Microbiology, Monash University, Clayton, Victoria, Australia, 3800
- School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China, 6009
| |
Collapse
|
30
|
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
|
31
|
Murai T, Matsuda S. Metabolic Reprogramming toward Aerobic Glycolysis and the Gut Microbiota Involved in the Brain Amyloid Pathology. BIOLOGY 2023; 12:1081. [PMID: 37626967 PMCID: PMC10452252 DOI: 10.3390/biology12081081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023]
Abstract
Alzheimer's disease (AD) is characterized by the formation of senile plaques consisting of fibrillated amyloid-β (Aβ), dystrophic neurites, and the neurofibrillary tangles of tau. The oligomers/fibrillar Aβ damages the neurons or initiates an intracellular signaling cascade for neuronal cell death leading to Aβ toxicity. The Aβ is a 4 kDa molecular weight peptide originating from the C-terminal region of the amyloid precursor protein via proteolytic cleavage. Apart from the typical AD hallmarks, certain deficits in metabolic alterations have been identified. This study describes the emerging features of AD from the aspect of metabolic reprogramming in the main pathway of carbohydrate metabolism in the human brain. Particularly, the neurons in patients with AD favor glycolysis despite a normal mitochondrial function indicating a Warburg-like effect. In addition, certain dietary patterns are well known for their properties in preventing AD. Among those, a ketogenic diet may substantially improve the symptoms of AD. An effective therapeutic method for the treatment, mitigation, and prevention of AD has not yet been established. Therefore, the researchers pursue the development and establishment of novel therapies effective in suppressing AD symptoms and the elucidation of their underlying protective mechanisms against neurodegeneration aiming for AD therapy in the near future.
Collapse
Affiliation(s)
- Toshiyuki Murai
- Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita 565-0871, Japan;
| | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| |
Collapse
|
32
|
Lin Y, Lin A, Cai L, Huang W, Yan S, Wei Y, Ruan X, Fang W, Dai X, Cheng J, Zhang J, Chen W, Ye Q, Chen X, Zhang J. ACSS2-dependent histone acetylation improves cognition in mouse model of Alzheimer's disease. Mol Neurodegener 2023; 18:47. [PMID: 37438762 DOI: 10.1186/s13024-023-00625-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 05/15/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Nuclear acetyl-CoA pools govern histone acetylation that controls synaptic plasticity and contributes to cognitive deterioration in patients with Alzheimer's disease (AD). Nuclear acetyl-CoA pools are generated partially from local acetate that is metabolized by acetyl-CoA synthetase 2 (ACSS2). However, the underlying mechanism of histone acetylation dysregulation in AD remains poorly understood. METHODS We detected ACSS2 expression and histone acetylation levels in the brains of AD patients and 5 × FAD mice. When we altered ACSS2 expression by injecting adeno-associated virus into the dorsal hippocampus of 5 × FAD mice and replenished ACSS2 substrate (acetate), we observed changes in cognitive function by Morris water maze. We next performed RNA-seq, ChIP-qPCR, and electrophysiology to study molecular mechanism underlying ACSS2-mediated spatial learning and memory in 5 × FAD mice. RESULTS We reported that ACSS2 expression and histone acetylation (H3K9, H4K12) were reduced in the hippocampus and prefrontal cortex of 5 × FAD mice. Reduced ACSS2 levels were also observed in the temporal cortex of AD patients. 5 × FAD mice exhibited a low enrichment of acetylated histones on the promoters of NMDARs and AMPARs, together with impaired basal and activity-dependent synaptic plasticity, all of which were rescued by ACSS2 upregulation. Moreover, acetate replenishment enhanced ac-H3K9 and ac-H4K12 in 5 × FAD mice, leading to an increase of NMDARs and AMPARs and a restoration of synaptic plasticity and cognitive function in an ACSS2-dependent manner. CONCLUSION ACSS2 is a key molecular switch of cognitive impairment and that targeting ACSS2 or acetate administration may serve as a novel therapeutic strategy for the treatment of intermediate or advanced AD. Nuclear acetyl-CoA pools are generated partly from local acetate that is metabolized by acetyl-CoA synthetase 2 (ACSS2). Model depicts that ACSS2 expression is downregulated in the brains of 5×FAD model mice and AD patients. Of note, ACSS2 downregulation mediates a reduction in ionotropic glutamate receptor expression through histone acetylation, which exacerbates synaptic plasticity impairment in AD. These deficits can be rescued by ACSS2 upregulation or acetate supplementation (GTA, an FDA-approved food additive), which may serve as a promising therapeutic strategy for AD treatment.
Collapse
Affiliation(s)
- Yingbin Lin
- Department of Neurology, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University, Fuzhou, China
- Department of Neurology and Neurosurgery, Institute of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Anlan Lin
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Lili Cai
- Department of Neurology, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Weibin Huang
- Department of Neurology, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University, Fuzhou, China
- Department of Neurology and Neurosurgery, Institute of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Shanzhi Yan
- Department of Neurology, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Yuanxiang Wei
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xinglin Ruan
- Department of Neurology, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Wenting Fang
- Department of Neurology, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Xiaoman Dai
- Department of Neurology, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Jinbo Cheng
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jie Zhang
- Department of Neurology, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Wanjin Chen
- Department of Neurology and Neurosurgery, Institute of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Qinyong Ye
- Department of Neurology, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Xiaochun Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University, Fuzhou, China.
| | - Jing Zhang
- Department of Neurology, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
33
|
Hartnett KB, Ferguson BJ, Hecht PM, Schuster LE, Shenker JI, Mehr DR, Fritsche KL, Belury MA, Scharre DW, Horwitz AJ, Kille BM, Sutton BE, Tatum PE, Greenlief CM, Beversdorf DQ. Potential Neuroprotective Effects of Dietary Omega-3 Fatty Acids on Stress in Alzheimer's Disease. Biomolecules 2023; 13:1096. [PMID: 37509132 PMCID: PMC10377362 DOI: 10.3390/biom13071096] [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: 05/26/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND A large number of individual potentially modifiable factors are associated with risk for Alzheimer's disease (AD). However, less is known about the interactions between the individual factors. METHODS In order to begin to examine the relationship between a pair of factors, we performed a pilot study, surveying patients with AD and controls for stress exposure and dietary omega-3 fatty acid intake to explore their relationship for risk of AD. RESULTS For individuals with the greatest stress exposure, omega-3 fatty acid intake was significantly greater in healthy controls than in AD patients. There was no difference among those with low stress exposure. CONCLUSIONS These initial results begin to suggest that omega-3 fatty acids may mitigate AD risk in the setting of greater stress exposure. This will need to be examined with larger populations and other pairs of risk factors to better understand these important relationships. Examining how individual risk factors interact will ultimately be important for learning how to optimally decrease the risk of AD.
Collapse
Affiliation(s)
- Kaitlyn B Hartnett
- School of Medicine, University of Missouri-Columbia, Columbia, MO 65212, USA
| | - Bradley J Ferguson
- Interdisciplinary Neuroscience Program, University of Missouri-Columbia, Columbia, MO 65212, USA
- Department of Health Psychology, University of Missouri-Columbia, Columbia, MO 65212, USA
- Department of Neurology, University of Missouri-Columbia, Columbia, MO 65212, USA
| | - Patrick M Hecht
- Interdisciplinary Neuroscience Program, University of Missouri-Columbia, Columbia, MO 65212, USA
| | - Luke E Schuster
- School of Medicine, University of Kansas, Kansas City, KS 66160, USA
| | - Joel I Shenker
- Department of Neurology, University of Missouri-Columbia, Columbia, MO 65212, USA
| | - David R Mehr
- Family & Community Medicine, University of Missouri-Columbia, Columbia, MO 65212, USA
| | - Kevin L Fritsche
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, USA
| | - Martha A Belury
- Department of Human Sciences, Ohio State University, Columbus, OH 43210, USA
| | - Douglas W Scharre
- Department of Neurology, Ohio State University, Columbus, OH 43210, USA
| | | | | | - Briann E Sutton
- College of Osteopathic Medicine, William Carey University, Hattiesburg, MS 39401, USA
| | - Paul E Tatum
- Division of Palliative Medicine; Washington University. St. Louis, MO 63110, USA
| | | | - David Q Beversdorf
- Interdisciplinary Neuroscience Program, University of Missouri-Columbia, Columbia, MO 65212, USA
- Department of Neurology, University of Missouri-Columbia, Columbia, MO 65212, USA
- Department of Radiology, University of Missouri, Columbia, MO 65212, USA
- Psychological Sciences, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
34
|
Fotuhi M, Khorrami ND, Raji CA. Benefits of a 12-Week Non-Drug "Brain Fitness Program" for Patients with Attention-Deficit/Hyperactive Disorder, Post-Concussion Syndrome, or Memory Loss. J Alzheimers Dis Rep 2023; 7:675-697. [PMID: 37483322 PMCID: PMC10357116 DOI: 10.3233/adr-220091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 05/25/2023] [Indexed: 07/25/2023] Open
Abstract
Background Non-pharmacologic interventions can potentially improve cognitive function, sleep, and/or mood in patients with attention-deficit/hyperactive disorder (ADHD), post-concussion syndrome (PCS), or memory loss. Objective We evaluated the benefits of a brain rehabilitation program in an outpatient neurology practice that consists of targeted cognitive training, lifestyle coaching, and electroencephalography (EEG)-based neurofeedback, twice weekly (90 minutes each), for 12 weeks. Methods 223 child and adult patients were included: 71 patients with ADHD, 88 with PCS, and 64 with memory loss (mild cognitive impairment or subjective cognitive decline). Patients underwent a complete neurocognitive evaluation, including tests for Verbal Memory, Complex Attention, Processing Speed, Executive Functioning, and Neurocognition Index. They completed questionnaires about sleep, mood, diet, exercise, anxiety levels, and depression-as well as underwent quantitative EEG-at the beginning and the end of the program. Results Pre-post test score comparison demonstrated that all patient subgroups experienced statistically significant improvements on most measures, especially the PCS subgroup, which experienced significant score improvement on all measures tested (p≤0.0011; dz≥0.36). After completing the program, 60% to 90% of patients scored higher on cognitive tests and reported having fewer cognitive and emotional symptoms. The largest effect size for pre-post score change was improved executive functioning in all subgroups (ADHD dz= 0.86; PCS dz= 0.83; memory dz= 1.09). Conclusion This study demonstrates that a multimodal brain rehabilitation program can have benefits for patients with ADHD, PCS, or memory loss and supports further clinical trials in this field.
Collapse
Affiliation(s)
- Majid Fotuhi
- Department of Psychological & Brain Sciences, George Washington University, Washington, DC, USA
- NeuroGrow Brain Fitness Center, McLean, VA, USA
| | | | - Cyrus A. Raji
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
35
|
Doroszkiewicz J, Mroczko J, Rutkowski P, Mroczko B. Molecular Aspects of a Diet as a New Pathway in the Prevention and Treatment of Alzheimer's Disease. Int J Mol Sci 2023; 24:10751. [PMID: 37445928 PMCID: PMC10341644 DOI: 10.3390/ijms241310751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Alzheimer's disease is the most common cause of dementia in the world. Lack of an established pathology makes it difficult to develop suitable approaches and treatment for the disease. Besides known hallmarks, including amyloid β peptides cumulating in plaques and hyperphosphorylated tau forming NFTs, inflammation also plays an important role, with known connections to the diet. In AD, adhering to reasonable nutrition according to age-related principles is recommended. The diet should be high in neuroprotective foods, such as polyunsaturated fatty acids, antioxidants, and B vitamins. In addition, foods capable of rising BDNF should be considered because of the known profitable results of this molecule in AD. Adhering to beneficial diets might result in improvements in memory, cognition, and biomarkers and might even reduce the risk of developing AD. In this review, we discuss the effects of various diets, foods, and nutrients on brain health and possible connections to Alzheimer's disease.
Collapse
Affiliation(s)
- Julia Doroszkiewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-089 Bialystok, Poland; (J.M.); (B.M.)
| | - Jan Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-089 Bialystok, Poland; (J.M.); (B.M.)
| | | | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-089 Bialystok, Poland; (J.M.); (B.M.)
- Department of Biochemical Diagnostics, Medical University of Białystok, 15-089 Bialystok, Poland
| |
Collapse
|
36
|
Reiss AB, Muhieddine D, Jacob B, Mesbah M, Pinkhasov A, Gomolin IH, Stecker MM, Wisniewski T, De Leon J. Alzheimer's Disease Treatment: The Search for a Breakthrough. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1084. [PMID: 37374288 PMCID: PMC10302500 DOI: 10.3390/medicina59061084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/22/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023]
Abstract
As the search for modalities to cure Alzheimer's disease (AD) has made slow progress, research has now turned to innovative pathways involving neural and peripheral inflammation and neuro-regeneration. Widely used AD treatments provide only symptomatic relief without changing the disease course. The recently FDA-approved anti-amyloid drugs, aducanumab and lecanemab, have demonstrated unclear real-world efficacy with a substantial side effect profile. Interest is growing in targeting the early stages of AD before irreversible pathologic changes so that cognitive function and neuronal viability can be preserved. Neuroinflammation is a fundamental feature of AD that involves complex relationships among cerebral immune cells and pro-inflammatory cytokines, which could be altered pharmacologically by AD therapy. Here, we provide an overview of the manipulations attempted in pre-clinical experiments. These include inhibition of microglial receptors, attenuation of inflammation and enhancement of toxin-clearing autophagy. In addition, modulation of the microbiome-brain-gut axis, dietary changes, and increased mental and physical exercise are under evaluation as ways to optimize brain health. As the scientific and medical communities work together, new solutions may be on the horizon to slow or halt AD progression.
Collapse
Affiliation(s)
- Allison B. Reiss
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| | - Dalia Muhieddine
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| | - Berlin Jacob
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| | - Michael Mesbah
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| | - Aaron Pinkhasov
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| | - Irving H. Gomolin
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| | | | - Thomas Wisniewski
- Center for Cognitive Neurology, Departments of Neurology, Pathology and Psychiatry, NYU School of Medicine, New York, NY 10016, USA;
| | - Joshua De Leon
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| |
Collapse
|
37
|
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: 40] [Impact Index Per Article: 40.0] [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
|
38
|
Thomas A, Ryan CP, Caspi A, Moffitt TE, Sugden K, Zhou J, Belsky DW, Gu Y. Diet, pace of biological aging, and risk of dementia in the Framingham Heart Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.24.23290474. [PMID: 37398353 PMCID: PMC10312831 DOI: 10.1101/2023.05.24.23290474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
INTRODUCTION We tested the hypothesis that healthy diet protects against dementia because it slows the pace of biological aging. METHODS We analyzed Framingham Offspring Cohort data (≥60y). We measured healthy diet using the Dietary Guideline for Americans (DGA, 3 visits 1991-2008), pace of aging using the DunedinPACE epigenetic clock (2005-2008), and incident dementia and mortality using records (compiled 2005-2018). RESULTS Of n=1,525 included participants (mean age 69.7, 54% female), n=129 developed dementia and n=432 died over follow-up. Greater DGA adherence was associated with slower DunedinPACE and reduced risks for dementia and mortality. Slower DunedinPACE was associated with reduced risks for dementia and mortality. Slower DunedinPACE accounted for 15% of the DGA association with dementia and 39% of the DGA association with mortality. DISCUSSION Findings suggest that slower pace of aging mediates part of the relationship of healthy diet with reduced dementia risk. Monitoring pace of aging may inform dementia prevention.
Collapse
Affiliation(s)
- Aline Thomas
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
| | - Calen P Ryan
- Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Avshalom Caspi
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Terrie E. Moffitt
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Karen Sugden
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Jiayi Zhou
- Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Daniel W. Belsky
- Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA
- Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Yian Gu
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
- Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University, New York, NY, USA
- Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA
| |
Collapse
|
39
|
Badaeva AV, Danilov AB, Clayton P, Moskalev AA, Karasev AV, Tarasevich AF, Vorobyeva YD, Novikov VN. Perspectives on Neuronutrition in Prevention and Treatment of Neurological Disorders. Nutrients 2023; 15:nu15112505. [PMID: 37299468 DOI: 10.3390/nu15112505] [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: 04/22/2023] [Revised: 05/16/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
The term neuronutrition has been proposed as part of nutritional neuroscience, studying the effects of various dietary components on behavior and cognition. Other researchers underline that neuronutrition includes the use of various nutrients and diets to prevent and treat neurological disorders. The aim of this narrative review was to explore the current understanding of the term neuronutrition as the key concept for brain health, its potential molecular targets, and perspectives of its nutritional approach to the prevention and treatment of Alzheimer's and Parkinson's diseases, multiple sclerosis, anxiety, depressive disorders, migraine, and chronic pain. Neuronutrition can be defined as a part of neuroscience that studies the influence of various aspects of nutrition (nutrients, diet, eating behavior, food environment, etc.) on the development of nervous disorders and includes nutrition, clinical dietetics, and neurology. There is evidence that the neuronutritional approach can influence neuroepigenetic modifications, immunological regulation, metabolic control, and behavioral patterns. The main molecular targets in neuronutrition include neuroinflammation, oxidative/nitrosative stress and mitochondrial dysfunction, gut-brain axis disturbance, and neurotransmitter imbalance. To effectively apply neuronutrition for maintaining brain health, a personalized approach is needed, which includes the adaptation of the scientific findings to the genetic, biochemical, psycho-physiological, and environmental features of each individual.
Collapse
Affiliation(s)
- Anastasiia V Badaeva
- Department of Personalized and Preventive Medicine, Institute of Interdisciplinary Medicine, 107113 Moscow, Russia
- Department for Nervous Diseases, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
| | - Alexey B Danilov
- Department of Personalized and Preventive Medicine, Institute of Interdisciplinary Medicine, 107113 Moscow, Russia
- Department for Nervous Diseases, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
| | - Paul Clayton
- Department of Personalized and Preventive Medicine, Institute of Interdisciplinary Medicine, 107113 Moscow, Russia
| | - Alexey A Moskalev
- Russian Research Clinical Center of Gerontology of the Russian National Research Medical University Named after N.I. Pirogov, 129226 Moscow, Russia
| | - Alexander V Karasev
- Department of Personalized and Preventive Medicine, Institute of Interdisciplinary Medicine, 107113 Moscow, Russia
| | - Andrey F Tarasevich
- Department of Personalized and Preventive Medicine, Institute of Interdisciplinary Medicine, 107113 Moscow, Russia
| | - Yulia D Vorobyeva
- Department for Nervous Diseases, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
| | - Viacheslav N Novikov
- Department for Nervous Diseases, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
| |
Collapse
|
40
|
Xu Lou I, Ali K, Chen Q. Effect of nutrition in Alzheimer's disease: A systematic review. Front Neurosci 2023; 17:1147177. [PMID: 37214392 PMCID: PMC10194838 DOI: 10.3389/fnins.2023.1147177] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/01/2023] [Indexed: 05/24/2023] Open
Abstract
Background and objective Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by declining cognitive ability. Currently, there are no effective treatments for this condition. However, certain measures, such as nutritional interventions, can slow disease progression. Therefore, the objective of this systematic review was to identify and map the updates of the last 5 years regarding the nutritional status and nutritional interventions associated with AD patients. Study design A systematic review. Methods A search was conducted for randomized clinical trials, systematic reviews, and meta-analyses investigating the association between nutritional interventions and AD published between 2018 and 2022 in the PubMed, Web of Science, Scopus, and Cochrane Library databases. A total of 38 studies were identified, of which 17 were randomized clinical trials, and 21 were systematic reviews and/or meta-analyses. Results The results show that the western diet pattern is a risk factor for developing AD. In contrast, the Mediterranean diet, ketogenic diet, and supplementation with omega-3 fatty acids and probiotics are protective factors. This effect is significant only in cases of mild-to-moderate AD. Conclusion Certain nutritional interventions may slow the progression of AD and improve cognitive function and quality of life. Further research is required to draw more definitive conclusions.
Collapse
Affiliation(s)
- Inmaculada Xu Lou
- International Education College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Department of Cardiology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Kamran Ali
- Department of Oncology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qilan Chen
- Department of Cardiology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
41
|
Abstract
Reviewing the research presented in this article, it is evident that from an epidemiological perspective, it is important to evaluate the extent to which findings of sex and gender differences in Alzheimer's dementia (AD) are due to differences in longevity, survival bias, and comorbidities. Medical, genetic, psychosocial, and behavioral factors, in addition to hormonal factors, can differentially affect the risk and progression of AD in women versus men. Further, evaluation of sex differences in AD progression and the trajectory of change in cognitive function, neuroimaging, cerebrospinal fluid (CSF), and blood-based biomarkers of AD is needed. Finally, identifying sex differences in AD biomarkers and change across the lifespan is critical for the planning of prevention trials to reduce the risk of developing AD.
Collapse
Affiliation(s)
- Neelum T Aggarwal
- Department of Neurological Sciences, Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 West Harrison Street, Suite 1000, Chicago, IL 60612, USA.
| | - Michelle M Mielke
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
42
|
Levakov G, Kaplan A, Yaskolka Meir A, Rinott E, Tsaban G, Zelicha H, Blüher M, Ceglarek U, Stumvoll M, Shelef I, Avidan G, Shai I. The effect of weight loss following 18 months of lifestyle intervention on brain age assessed with resting-state functional connectivity. eLife 2023; 12:e83604. [PMID: 37022140 PMCID: PMC10174688 DOI: 10.7554/elife.83604] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
Background Obesity negatively impacts multiple bodily systems, including the central nervous system. Retrospective studies that estimated chronological age from neuroimaging have found accelerated brain aging in obesity, but it is unclear how this estimation would be affected by weight loss following a lifestyle intervention. Methods In a sub-study of 102 participants of the Dietary Intervention Randomized Controlled Trial Polyphenols Unprocessed Study (DIRECT-PLUS) trial, we tested the effect of weight loss following 18 months of lifestyle intervention on predicted brain age based on magnetic resonance imaging (MRI)-assessed resting-state functional connectivity (RSFC). We further examined how dynamics in multiple health factors, including anthropometric measurements, blood biomarkers, and fat deposition, can account for changes in brain age. Results To establish our method, we first demonstrated that our model could successfully predict chronological age from RSFC in three cohorts (n=291;358;102). We then found that among the DIRECT-PLUS participants, 1% of body weight loss resulted in an 8.9 months' attenuation of brain age. Attenuation of brain age was significantly associated with improved liver biomarkers, decreased liver fat, and visceral and deep subcutaneous adipose tissues after 18 months of intervention. Finally, we showed that lower consumption of processed food, sweets and beverages were associated with attenuated brain age. Conclusions Successful weight loss following lifestyle intervention might have a beneficial effect on the trajectory of brain aging. Funding The German Research Foundation (DFG), German Research Foundation - project number 209933838 - SFB 1052; B11, Israel Ministry of Health grant 87472511 (to I Shai); Israel Ministry of Science and Technology grant 3-13604 (to I Shai); and the California Walnuts Commission 09933838 SFB 105 (to I Shai).
Collapse
Affiliation(s)
- Gidon Levakov
- Department of Brain and Cognitive Sciences, Ben-Gurion University of the NegevBeer ShevaIsrael
| | - Alon Kaplan
- The Health & Nutrition Innovative International Research Center, Faculty of Health Sciences, Ben-Gurion University of the NegevBeer ShevaIsrael
- Department of Internal Medicine D, Chaim Sheba Medical CenterRamat-GanIsrael
| | - Anat Yaskolka Meir
- The Health & Nutrition Innovative International Research Center, Faculty of Health Sciences, Ben-Gurion University of the NegevBeer ShevaIsrael
| | - Ehud Rinott
- The Health & Nutrition Innovative International Research Center, Faculty of Health Sciences, Ben-Gurion University of the NegevBeer ShevaIsrael
| | - Gal Tsaban
- The Health & Nutrition Innovative International Research Center, Faculty of Health Sciences, Ben-Gurion University of the NegevBeer ShevaIsrael
| | - Hila Zelicha
- The Health & Nutrition Innovative International Research Center, Faculty of Health Sciences, Ben-Gurion University of the NegevBeer ShevaIsrael
| | | | - Uta Ceglarek
- Department of Medicine, University of LeipzigLeipzigGermany
| | | | - Ilan Shelef
- Department of Diagnostic Imaging, Soroka Medical CenterBeer ShevaIsrael
| | - Galia Avidan
- Department of Psychology, Ben-Gurion University of the NegevBeer ShevaIsrael
| | - Iris Shai
- The Health & Nutrition Innovative International Research Center, Faculty of Health Sciences, Ben-Gurion University of the NegevBeer ShevaIsrael
- Department of Medicine, University of LeipzigLeipzigGermany
- Department of Nutrition, Harvard T.H. Chan School of Public HealthBostonUnited States
| |
Collapse
|
43
|
Duggan MR, Butler L, Peng Z, Daya GN, Moghekar A, An Y, Rapp SR, Hayden KM, Shadyab AH, Natale G, Liu L, Snetselaar L, Moaddel R, Rebholz CM, Sullivan K, Ballantyne CM, Resnick SM, Ferrucci L, Walker KA. Plasma proteins related to inflammatory diet predict future cognitive impairment. Mol Psychiatry 2023; 28:1599-1609. [PMID: 36737481 PMCID: PMC10208977 DOI: 10.1038/s41380-023-01975-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 02/05/2023]
Abstract
Dysregulation of the immune system and dietary patterns that increase inflammation can increase the risk for cognitive decline, but the mechanisms by which inflammatory nutritional habits may affect the development of cognitive impairment in aging are not well understood. To determine whether plasma proteins linked to inflammatory diet predict future cognitive impairment, we applied high-throughput proteomic assays to plasma samples from a subset (n = 1528) of Women's Health Initiative Memory Study (WHIMS) participants (mean [SD] baseline age, 71.3 [SD 3.8] years). Results provide insights into how inflammatory nutritional patterns are associated with an immune-related proteome and identify a group of proteins (CXCL10, CCL3, HGF, OPG, CDCP1, NFATC3, ITGA11) related to future cognitive impairment over a 14-year follow-up period. Several of these inflammatory diet proteins were also associated with dementia risk across two external cohorts (ARIC, ESTHER), correlated with plasma biomarkers of Alzheimer's disease (AD) pathology (Aβ42/40) and/or neurodegeneration (NfL), and related to an MRI-defined index of neurodegenerative brain atrophy in a separate cohort (BLSA). In addition to evaluating their biological relevance, assessing their potential role in AD, and characterizing their immune-tissue/cell-specific expression, we leveraged published RNA-seq results to examine how the in vitro regulation of genes encoding these candidate proteins might be altered in response to an immune challenge. Our findings indicate how dietary patterns with higher inflammatory potential relate to plasma levels of immunologically relevant proteins and highlight the molecular mediators which predict subsequent risk for age-related cognitive impairment.
Collapse
Affiliation(s)
- Michael R Duggan
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Lauren Butler
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Zhongsheng Peng
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Gulzar N Daya
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Stephen R Rapp
- Department of Social Sciences and Health Policy, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Psychiatry & Behavioral Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Kathleen M Hayden
- Department of Psychiatry & Behavioral Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Aladdin H Shadyab
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA, USA
| | - Ginny Natale
- Program in Public Health, Stony Brook University School of Medicine, Stony Brook, NY, USA
| | - Longjian Liu
- Department of Epidemiology and Biostatistics, Drexel University Dornsife School of Public Health, Philadelphia, PA, USA
| | - Linda Snetselaar
- Department of Epidemiology, University of Iowa, Iowa City, IA, USA
| | - Ruin Moaddel
- Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Casey M Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kevin Sullivan
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Christie M Ballantyne
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA.
| |
Collapse
|
44
|
Abstract
This article describes the public health impact of Alzheimer's disease, including prevalence and incidence, mortality and morbidity, use and costs of care, and the overall impact on family caregivers, the dementia workforce and society. The Special Report examines the patient journey from awareness of cognitive changes to potential treatment with drugs that change the underlying biology of Alzheimer's. An estimated 6.7 million Americans age 65 and older are living with Alzheimer's dementia today. This number could grow to 13.8 million by 2060 barring the development of medical breakthroughs to prevent, slow or cure AD. Official death certificates recorded 121,499 deaths from AD in 2019, and Alzheimer's disease was officially listed as the sixth-leading cause of death in the United States. In 2020 and 2021, when COVID-19 entered the ranks of the top ten causes of death, Alzheimer's was the seventh-leading cause of death. Alzheimer's remains the fifth-leading cause of death among Americans age 65 and older. Between 2000 and 2019, deaths from stroke, heart disease and HIV decreased, whereas reported deaths from AD increased more than 145%. This trajectory of deaths from AD was likely exacerbated by the COVID-19 pandemic in 2020 and 2021. More than 11 million family members and other unpaid caregivers provided an estimated 18 billion hours of care to people with Alzheimer's or other dementias in 2022. These figures reflect a decline in the number of caregivers compared with a decade earlier, as well as an increase in the amount of care provided by each remaining caregiver. Unpaid dementia caregiving was valued at $339.5 billion in 2022. Its costs, however, extend to family caregivers' increased risk for emotional distress and negative mental and physical health outcomes - costs that have been aggravated by COVID-19. Members of the paid health care workforce are involved in diagnosing, treating and caring for people with dementia. In recent years, however, a shortage of such workers has developed in the United States. This shortage - brought about, in part, by COVID-19 - has occurred at a time when more members of the dementia care workforce are needed. Therefore, programs will be needed to attract workers and better train health care teams. Average per-person Medicare payments for services to beneficiaries age 65 and older with AD or other dementias are almost three times as great as payments for beneficiaries without these conditions, and Medicaid payments are more than 22 times as great. Total payments in 2023 for health care, long-term care and hospice services for people age 65 and older with dementia are estimated to be $345 billion. The Special Report examines whether there will be sufficient numbers of physician specialists to provide Alzheimer's care and treatment now that two drugs are available that change the underlying biology of Alzheimer's disease.
Collapse
|
45
|
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: 4.0] [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
|
46
|
Tsiknia AA, Bergstrom J, Reas ET. Midlife omega-3 fatty acid intake predicts later life white matter microstructure in an age- and APOE-dependent manner. Cereb Cortex 2023; 33:2143-2151. [PMID: 35584792 PMCID: PMC9977375 DOI: 10.1093/cercor/bhac196] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Omega-3 intake has been positively associated with healthy brain aging, yet it remains unclear whether high omega-3 intake beginning early in life may optimize its protective effects against brain aging. We examined whether omega-3 intake is associated with brain microstructure over 2 decades later among dementia-free older adults. The 128 participants (62% women; age at magnetic resonance imaging: 76.6 ± 7.9) from the Rancho Bernardo Study of Healthy Aging completed at least 1 dietary assessment between 1984 and 1996 and underwent restriction spectrum imaging (RSI) 22.8 ± 3.1 years later. We evaluated associations between prior omega-3 intake and RSI metrics of gray and white matter (WM) microstructure. Higher prior omega-3 intake was associated with greater restricted diffusion in the superior cortico-striatal fasciculus. A correlation between higher prior omega-3 intake and greater cingulum restricted diffusion was stronger among participants >80 years old. Higher omega-3 intake correlated with greater restricted diffusion in the inferior longitudinal and inferior fronto-occipital fasciculus more strongly for apolipoprotein E (APOE) ε4 carriers than noncarriers. Associations were not modified by adjustment for dietary pattern, health, or lifestyle. High omega-3 intake in midlife may help to maintain WM integrity into older age, particularly in the latest decades of life and among APOE ε4 carriers.
Collapse
Affiliation(s)
- Amaryllis A Tsiknia
- Department of Neurosciences, University of California, San Diego, CA 92093-0841, United States
| | - Jaclyn Bergstrom
- School of Public Health and Human Longevity Science, University of California, San Diego, CA 92093, United States
| | - Emilie T Reas
- Department of Neurosciences, University of California, San Diego, CA 92093-0841, United States
| |
Collapse
|
47
|
Chandra S, Sisodia SS, Vassar RJ. The gut microbiome in Alzheimer's disease: what we know and what remains to be explored. Mol Neurodegener 2023; 18:9. [PMID: 36721148 PMCID: PMC9889249 DOI: 10.1186/s13024-023-00595-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/06/2023] [Indexed: 02/02/2023] Open
Abstract
Alzheimer's disease (AD), the most common cause of dementia, results in a sustained decline in cognition. There are currently few effective disease modifying therapies for AD, but insights into the mechanisms that mediate the onset and progression of disease may lead to new, effective therapeutic strategies. Amyloid beta oligomers and plaques, tau aggregates, and neuroinflammation play a critical role in neurodegeneration and impact clinical AD progression. The upstream modulators of these pathological features have not been fully clarified, but recent evidence indicates that the gut microbiome (GMB) may have an influence on these features and therefore may influence AD progression in human patients. In this review, we summarize studies that have identified alterations in the GMB that correlate with pathophysiology in AD patients and AD mouse models. Additionally, we discuss findings with GMB manipulations in AD models and potential GMB-targeted therapeutics for AD. Lastly, we discuss diet, sleep, and exercise as potential modifiers of the relationship between the GMB and AD and conclude with future directions and recommendations for further studies of this topic.
Collapse
Affiliation(s)
- Sidhanth Chandra
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
- Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Sangram S. Sisodia
- Department of Neurobiology, University of Chicago, Chicago, IL 60637 USA
| | - Robert J. Vassar
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| |
Collapse
|
48
|
Katz Sand I, Levy S, Fitzgerald K, Sorets T, Sumowski JF. Mediterranean diet is linked to less objective disability in multiple sclerosis. Mult Scler 2023; 29:248-260. [PMID: 36226971 PMCID: PMC9918647 DOI: 10.1177/13524585221127414] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The multiple sclerosis (MS) community is highly interested in diet as a potential protective factor against disability, but empirical evidence remains limited. OBJECTIVE Evaluate associations between patient-reported Mediterranean diet alignment and objective disability in a real-world MS cohort. METHODS Data were analyzed from persons with MS, aged 18-65, who completed the Mediterranean Diet Adherence Screener (MEDAS), MS Functional Composite (MSFC; primary disability metric), and patient-reported outcomes (PROs; disability, gait disturbance, fatigue, anxiety, and depression) as part of our Comprehensive Annual Assessment Program. Multiple regression predicted MSFC (and PROs) with MEDAS after adjusting for demographic (age, sex, race, ethnicity, and socioeconomic status) and health-related (body mass index (BMI), exercise, sleep disturbance, hypertension, diabetes, hyperlipidemia, and smoking) covariates. RESULTS Higher MEDAS independently predicted better outcomes across MSFC (z-score, B = 0.10 (95% confidence interval (CI): 0.06, 0.13), β = 0.18, p < 0.001), MSFC components, and PROs in 563 consecutive patients. Each MEDAS point was associated with 15.0% lower risk for MSFC impairment (⩽ 5th percentile on ⩾ 2 tasks; odds ratio (OR) = 0.850; 95% CI: 0.779, 0.928). Higher MEDAS attenuated effects of progressive disease and longer disease duration on disability. CONCLUSION With robust control for potential confounds, higher Mediterranean diet alignment predicted lower objective and patient-reported disability. Findings lay the necessary groundwork for longitudinal and interventional studies to guide clinical recommendations in MS.
Collapse
Affiliation(s)
- Ilana Katz Sand
- Department of Neurology, Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah Levy
- Department of Neurology, Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Tali Sorets
- Department of Neurology, Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James F Sumowski
- Department of Neurology, Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
49
|
Thwarting Alzheimer's Disease through Healthy Lifestyle Habits: Hope for the Future. Neurol Int 2023; 15:162-187. [PMID: 36810468 PMCID: PMC9944470 DOI: 10.3390/neurolint15010013] [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: 11/29/2022] [Revised: 12/17/2022] [Accepted: 12/26/2022] [Indexed: 01/31/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that slowly disintegrates memory and thinking skills. Age is known to be the major risk factor in AD, but there are several nonmodifiable and modifiable causes. The nonmodifiable risk factors such as family history, high cholesterol, head injuries, gender, pollution, and genetic aberrations are reported to expediate disease progression. The modifiable risk factors of AD that may help prevent or delay the onset of AD in liable people, which this review focuses on, includes lifestyle, diet, substance use, lack of physical and mental activity, social life, sleep, among other causes. We also discuss how mitigating underlying conditions such as hearing loss and cardiovascular complications could be beneficial in preventing cognitive decline. As the current medications can only treat the manifestations of AD and not the underlying process, healthy lifestyle choices associated with modifiable factors is the best alternative strategy to combat the disease.
Collapse
|
50
|
Grao-Cruces E, Calvo JR, Maldonado-Aibar MD, Millan-Linares MDC, Montserrat-de la Paz S. Mediterranean Diet and Melatonin: A Systematic Review. Antioxidants (Basel) 2023; 12:264. [PMID: 36829823 PMCID: PMC9951922 DOI: 10.3390/antiox12020264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
The Mediterranean diet (MD) has beneficial effects on human health, which is evidenced by the observation of lower incidence rates of chronic diseases in Mediterranean countries. The MD dietary pattern is rich in antioxidants, such as melatonin, which is a hormone produced mainly by the pineal gland and controls several circadian rhythms. Additionally, melatonin is found in foods, such as fruit and vegetables. The purpose of this systematic review was to assess the melatonin content in Mediterranean foods and to evaluate the influence of the MD on melatonin levels in both humans and model organisms. A comprehensive search was conducted in four databases (PubMed, Scopus, Cochrane Library and Web of Science) and data were extracted. A total of 31 records were chosen. MD-related foods, such as tomatoes, olive oil, red wine, beer, nuts, and vegetables, showed high melatonin contents. The consumption of specific MD foods increases melatonin levels and improves the antioxidant status in plasma.
Collapse
Affiliation(s)
| | | | | | | | - Sergio Montserrat-de la Paz
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Avenida Sanchez Pizjuan s/n, 41009 Seville, Spain
| |
Collapse
|