1
|
Yavari M, Kalupahana NS, Harris BN, Ramalingam L, Zu Y, Kahathuduwa CN, Moustaid-Moussa N. Mechanisms Linking Obesity, Insulin Resistance, and Alzheimer's Disease: Effects of Polyphenols and Omega-3 Polyunsaturated Fatty Acids. Nutrients 2025; 17:1203. [PMID: 40218960 PMCID: PMC11990358 DOI: 10.3390/nu17071203] [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: 03/05/2025] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized by progressive cognitive decline, memory loss, and behavioral changes. It poses a significant global health challenge. AD is associated with the accumulation of amyloid-β (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain, along with chronic inflammation, dysfunctional neurons, and synapse loss. While the prevalence of AD continues to rise, the current FDA-approved drugs offer only limited effectiveness. Emerging evidence suggests that obesity, insulin resistance (IR), and type 2 diabetes mellitus (T2DM) are also implicated in AD pathogenesis, with epidemiological studies and animal models confirming the impact of IR on Aβ accumulation, and high-fat diets also exacerbating Aβ accumulation. Since neuroinflammation activated by Aβ involves the nuclear factor kappa-light-chain-enhancer of the activated B cell (NF-κB) pathway, the inhibition of NF-κB and NLRP3 inflammasome activation are potential therapeutic strategies in AD. Bioactive compounds, including polyphenols (resveratrol, epigallocatechin-3-gallate, curcumin, and quercetin), and omega-3 polyunsaturated fatty acids, show promising results in animal studies and clinical trials for reducing Aβ levels, improving cognition and modulating the signaling pathways implicated in AD. This review explores the interplay between obesity, IR, inflammation, and AD pathology, emphasizing the potential of dietary compounds and their role in reducing inflammation, oxidative stress, and cognitive decline, as viable strategies for AD prevention and treatment. By integrating epidemiological findings, observational studies, and clinical trials, this review aims to provide a comprehensive understating of how metabolic dysfunctions and bioactive compounds influence AD progression.
Collapse
Affiliation(s)
- Mahsa Yavari
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; (M.Y.); (L.R.); (Y.Z.)
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
| | - Nishan Sudheera Kalupahana
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Breanna N. Harris
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Institute for One Health Innovation, Offices of Research & Innovation, Texas Tech University, Texas Tech Health Sciences Center, Lubbock, TX 79409, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; (M.Y.); (L.R.); (Y.Z.)
| | - Yujiao Zu
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; (M.Y.); (L.R.); (Y.Z.)
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
| | - Chanaka Nadeeshan Kahathuduwa
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
- Department of Neurology, Texas Tech University Health Sciences Center, El Paso, TX 79409, USA
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; (M.Y.); (L.R.); (Y.Z.)
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
- Institute for One Health Innovation, Offices of Research & Innovation, Texas Tech University, Texas Tech Health Sciences Center, Lubbock, TX 79409, USA
| |
Collapse
|
2
|
Tian A, Zheng Y, Li H, Zhang Z, Du L, Huang X, Sun L, Wu H. Eicosapentaenoic acid activates the P62/KEAP1/NRF2 pathway for the prevention of diabetes-associated cognitive dysfunction. Food Funct 2024; 15:5251-5271. [PMID: 38680120 DOI: 10.1039/d4fo00774c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Diabetes-associated cognitive dysfunction (DCD) is a severe complication of diabetes mellitus (DM), threatening the life quality of the diabetic population. However, there is still a lack of effective approaches for its intervention. Eicosapentaenoic acid (EPA) is an omega-3 polyunsaturated fatty acid that was not previously investigated for its effect on DCD. In this study, EPA was found to improve DCD in a mouse model of type 2 DM (T2DM) induced by streptozotocin and a high-fat diet, exhibiting profound protective effects on cognitive dysfunction, neuronal loss, and cerebral oxidative stress and inflammation. While EPA did not attenuate advanced glycation end product-induced neuron injury, we hypothesized that EPA might protect neurons by regulating microglia polarization, the effect of which was confirmed by the co-culture of neurons and lipopolysaccharide-stimulated microglia. RNA sequencing identified nuclear factor-erythroid-2-related factor 2 (NRF2) antioxidant signaling as a major target of EPA in microglia. Mechanistically, EPA increased sequestosome-1 (SQSTM1 or P62) levels that might structurally inhibit Kelch-like ECH associated protein 1 (KEAP1), leading to nuclear translocation of NRF2. P62 and NRF2 predominantly mediated EPA's effect since the knockdown of P62 or NRF2 abolished EPA's protective effect on microglial oxidative stress and inflammation and sequential neuron injuries. Moreover, the regulation of P62/KEPA1/NRF2 axes by EPA was confirmed in the hippocampi of diabetic mice. The present work presents EPA as an effective nutritional approach and microglial P62/KEAP1/NRF2 as molecular targets for the intervention of DCD.
Collapse
Affiliation(s)
- Ao Tian
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China.
| | - Yan Zheng
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
| | - Hui Li
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
| | - Zhiyue Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
| | - Lei Du
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China.
| | - Xiaoli Huang
- Department of Nutrition, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Rd., Jinan, Shandong 250012, China.
| | - Lei Sun
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Rd., Jinan, Shandong 250012, China.
| | - Hao Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China.
| |
Collapse
|
3
|
Harris BN, Yavari M, Ramalingam L, Mounce PL, Alers Maldonado K, Chavira AC, Thomas S, Scoggin S, Biltz C, Moustaid-Moussa N. Impact of Long-Term Dietary High Fat and Eicosapentaenoic Acid on Behavior and Hypothalamic-Pituitary-Adrenal Axis Activity in Amyloidogenic APPswe/PSEN1dE9 Mice. Neuroendocrinology 2024; 114:553-576. [PMID: 38301617 PMCID: PMC11153005 DOI: 10.1159/000536586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 01/30/2024] [Indexed: 02/03/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) alters neurocognitive and emotional function and causes dysregulation of multiple homeostatic processes. The leading AD framework pins amyloid beta plaques and tau tangles as primary drivers of dysfunction. However, many additional variables, including diet, stress, sex, age, and pain tolerance, interact in ways that are not fully understood to impact the onset and progression of AD pathophysiology. We asked: (1) does high-fat diet, compared to low-fat diet, exacerbate AD pathophysiology and behavioral decline? And, (2) can supplementation with eicosapentaenoic (EPA)-enriched fish oil prevent high-fat-diet-induced changes? METHODS Male and female APPswePSdE9 mice, and their non-transgenic littermates, were randomly assigned to a diet condition (low-fat, high-fat, high-fat with EPA) and followed from 2 to 10 months of age. We assessed baseline corticosterone concentration during aging, pain tolerance, cognitive function, stress coping, and corticosterone response to a stressor. RESULTS Transgenic mice were consistently more active than non-transgenic mice but did not perform worse on either cognitive task, even though we recently reported that these same transgenic mice exhibited metabolic changes and had increased amyloid beta. Mice fed high-fat diet had higher baseline and post-stressor corticosterone, but diet did not impact cognition or pain tolerance. Sex had the biggest influence, as female mice were consistently more active and had higher corticosterone than males. CONCLUSION Overall, diet, genotype, and sex did not have consistent impacts on outcomes. We found little support for predicted interactions and correlations, suggesting diet impacts metabolic function and amyloid beta levels, but these outcomes do not translate to changes in behaviors measured here.
Collapse
Affiliation(s)
- Breanna N. Harris
- Department of Biological Sciences, Texas Tech University, Lubbock, TX
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University
| | - Mahsa Yavari
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University
- Current address: Department of Molecular Metabolism, School of Public Health, Harvard University, Boston, MA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University
- Current address: Department of Nutritional and Food Studies Syracuse University, Syracuse, NY
| | - P. Logan Mounce
- Department of Biological Sciences, Texas Tech University, Lubbock, TX
| | | | - Angela C. Chavira
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX
| | - Sarah Thomas
- Department of Biological Sciences, Texas Tech University, Lubbock, TX
| | - Shane Scoggin
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX
| | - Caroline Biltz
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University
| |
Collapse
|
4
|
Yavari M, Ramalingam L, Harris BN, Kahathuduwa CN, Chavira A, Biltz C, Mounce L, Maldonado KA, Scoggin S, Zu Y, Kalupahana NS, Yosofvand M, Moussa H, Moustaid-Moussa N. Eicosapentaenoic Acid Protects against Metabolic Impairments in the APPswe/PS1dE9 Alzheimer's Disease Mouse Model. J Nutr 2023; 153:1038-1051. [PMID: 36781072 PMCID: PMC10273166 DOI: 10.1016/j.tjnut.2023.01.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 01/18/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by amyloid-β (Aβ) plaques. Systemic inflammation and obesity may exacerbate AD pathogenesis. We previously reported anti-inflammatory and anti-obesity effects of EPA in mice. OBJECTIVES We aimed to determine whether EPA reduces obesity-associated metabolic dysfunctions and Aβ accumulation in AD amyloidogenic mice. METHODS Two-mo-old APPswe/PS1dE9 transgenic (TG) mice and non-TG littermates were randomly assigned to low fat (LF; 10% kcal fat), high fat (HF; 45% kcal fat), or EPA (36 g/kg)-supplemented HF diets. Body composition, glucose tolerance, and energy expenditure were measured, and serum and brain metabolic markers were tested 38 wk postintervention. Outcomes were statistically analyzed via 3-factor ANOVA, modeling genotype, sex, and diet interactions. RESULTS HF-fed males gained more weight than females (Δ = 61 mg; P < 0.001). Compared with LF, HF increased body weights of wild-type (WT) males (Δ = 31 mg; P < 0.001). EPA reduced HF-induced weight gain in WT males (Δ = 24 mg; P = 0.054) but not in females. HF mice showed decreased glucose clearance and respiratory energy compared with LF-fed groups (Δ = -1.31 g/dL; P < 0.001), with no significant effects of EPA. However, EPA conferred metabolic improvements by decreasing serum leptin and insulin (Δ = -2.51 g/mL and Δ = -0.694 ng/mL, respectively compared with HF, P ≤ 0.05) and increasing adiponectin (Δ = 21.6 ng/mL; P < 0.001). As we expected, TG mice expressed higher serum and brain Aβ than WT mice (Δ = 0.131 ng/mL; P < 0.001 and Δ = 0.56%; P < 0.01, respectively), and EPA reduced serum Aβ1-40 in TG males compared with HF (Δ = 0.053 ng/mL; P ≤ 0.05). CONCLUSIONS To our knowledge, this is the first report that EPA reduces serum Aβ1-40 in obese AD male mice, warranting further investigations into tissue-specific mechanisms of EPA in AD.
Collapse
Affiliation(s)
- Mahsa Yavari
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA; Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Breanna N Harris
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX, USA; Department of Biological Sciences, Texas Tech University, Lubbock, TX, USA
| | - Chanaka Nadeeshan Kahathuduwa
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX, USA; Department of Laboratory Science and Primary Care, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Angela Chavira
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Caroline Biltz
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Logan Mounce
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, USA
| | | | - Shane Scoggin
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Yujiao Zu
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA; Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX, USA
| | - Nishan Sudheera Kalupahana
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA; Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX, USA; Department of Physiology, University of Peradeniya, Sri Lanka
| | - Mohammad Yosofvand
- Department of Mechanical Engineering, Texas Tech University, Lubbock, TX, USA
| | - Hanna Moussa
- Department of Mechanical Engineering, Texas Tech University, Lubbock, TX, USA
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA; Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX, USA.
| |
Collapse
|
5
|
The effects of multi-nutrient formulas containing a combination of n-3 PUFA and B vitamins on cognition in the older adult: a systematic review and meta-analysis. Br J Nutr 2023; 129:428-441. [PMID: 35473808 PMCID: PMC9876812 DOI: 10.1017/s0007114522001283] [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] [Indexed: 02/04/2023]
Abstract
There is now evidence to suggest that there may be an interaction between B vitamins and n-3 PUFA, with suggestions that increasing intake of both nutrients simultaneously may benefit cognition in older adults. The aim of this systematic review was to investigate whether supplementation with a combination of n-3 PUFA and B vitamins can prevent cognitive decline in older adults. Randomised controlled trials conducted in older adults that measured cognitive function were retrieved. The included trials provided a combination of n-3 PUFA and B vitamins alone, or in combination with other nutrients. Trials that provided n-3 PUFA alone and also measured B vitamin status or provided B vitamin supplementation alone and measured n-3 PUFA status were also included. The databases searched were The Cochrane Library, EMBASE, CINAHL, Scopus and MEDLINE. A total of 14 papers were included in the analysis (n 4913; age: 60-70 years; follow-up 24 weeks to 4 years). The meta-analysis results found a significant benefit of nutrient formulas, which included both n-3 PUFA and B vitamins alongside other nutrients, v. placebo on global cognition assessed using composite scores from a neuropsychological test battery (G = 0·23, P = 0·002), global cognition using single measures of cognition (G = 0·28, P = 0·004) and episodic memory (G = 0·32, P = 0·001). The results indicate that providing a combination of n-3 PUFA and B vitamins as part of a multi-nutrient formula benefits cognition in older adults v. a placebo, and the potential for an interaction between these key nutrients should be considered in future experimental work.
Collapse
|
6
|
Babazadeh A, Vahed FM, Liu Q, Siddiqui SA, Kharazmi MS, Jafari SM. Natural Bioactive Molecules as Neuromedicines for the Treatment/Prevention of Neurodegenerative Diseases. ACS OMEGA 2023; 8:3667-3683. [PMID: 36743024 PMCID: PMC9893457 DOI: 10.1021/acsomega.2c06098] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/29/2022] [Indexed: 06/18/2023]
Abstract
The brain is vulnerable to different types of stresses, particularly oxidative stress as a result of oxygen requirements/utilization in the body. Large amounts of unsaturated fatty acids present in the brain increase this vulnerability. Neurodegenerative diseases (NDDs) are brain disorders that are characterized by the gradual loss of specific neurons and are attributed to broad evidence of cell-level oxidative stress. The accurate characterization of neurological disorders relies on several parameters along with genetics and environmental risk factors, making therapies less efficient to fight NDDs. On the way to tackle oxidative damage and discover efficient and safe therapies, bioactives are at the edge of NDD science. Naturally occurring bioactive compounds such as polyphenols, carotenoids, essential fatty acids, phytosterols, essential oils, etc. are particularly of interest owing to their potent antioxidant and anti-inflammatory activities, and they offer lots of brain-health-promoting features. This Review focuses on probing the neuroefficacy and bioefficacy of bioactives and their role in supporting relatively low antioxidative and low regenerative capacities of the brain, neurogenesis, neuroprotection, and ameliorating/treating NDDs.
Collapse
Affiliation(s)
- Afshin Babazadeh
- Center
for Motor Neuron Disease Research, Macquarie Medical School, Faculty
of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Fereshteh Mohammadi Vahed
- Center
for Motor Neuron Disease Research, Macquarie Medical School, Faculty
of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Qi Liu
- Institute
of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
- Jiangsu
Key Laboratory of Integrated Traditional Chinese and Western Medicine
for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Shahida Anusha Siddiqui
- Technical
University of Munich Campus Straubing for Biotechnology and Sustainability, Essigberg 3, 94315 Straubing, Germany
- German
Institute of Food Technologies (DIL e.V.), Prof.-von-Klitzing-Straße 7, 49610 D Quakenbrück, Germany
| | | | - Seid Mahdi Jafari
- Department
of Food Materials and Process Design Engineering, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan 4913815739, Iran
- Nutrition
and Bromatology Group, Department of Analytical Chemistry and Food
Science, Faculty of Science, Universidade
de Vigo, E-32004 Ourense, Spain
- College
of Food Science and Technology, Hebei Agricultural
University, Baoding 071001, China
| |
Collapse
|
7
|
Refaey MS, Shah MA, Fayed MA, Rasul A, Siddiqui MF, Qasim M, Althobaiti NA, Saleem U, Malik A, Blundell R, Eldahshan OA. Neuroprotective effects of steroids. PHYTONUTRIENTS AND NEUROLOGICAL DISORDERS 2023:283-304. [DOI: 10.1016/b978-0-12-824467-8.00005-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
8
|
Jie F, Yang X, Yang B, Liu Y, Wu L, Lu B. Stigmasterol attenuates inflammatory response of microglia via NF-κB and NLRP3 signaling by AMPK activation. Biomed Pharmacother 2022; 153:113317. [DOI: 10.1016/j.biopha.2022.113317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 11/02/2022] Open
|
9
|
Ponce J, Ulu A, Hanson C, Cameron-Smith E, Bertoni J, Wuebker J, Fisher A, Siu KC, Marmelat V, Adamec J, Bhatti D. Role of Specialized Pro-resolving Mediators in Reducing Neuroinflammation in Neurodegenerative Disorders. Front Aging Neurosci 2022; 14:780811. [PMID: 35250536 PMCID: PMC8891627 DOI: 10.3389/fnagi.2022.780811] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 01/21/2022] [Indexed: 12/30/2022] Open
Abstract
Alzheimer’s disease (AD) and Parkinson’s disease (PD) are neurodegenerative disorders that affect millions of individuals worldwide. As incidence of these conditions increases with age, there will undoubtedly be an increased prevalence of cases in the near future. Neuroinflammation is a hallmark in the development and progression of neurodegenerative diseases and prevention or resolution of chronic neuroinflammation may represent a novel approach to treatment. The present review highlights the potential of the anti-inflammatory and pro-resolving effects of polyunsaturated fatty acid (PUFA)-derived mediators (Specialized Pro-resolving Mediators—SPM) in neurodegenerative disorders. PUFA-derived SPM are biosynthesized in response to chemicals produced from acute inflammatory responses. Preclinical studies from both AD and PD models suggest a dysregulation of SPM and their receptors in neurological disorders. Decreased SPM may be due to inadequate substrate, an imbalance between SPM and pro-inflammatory mediators or a disruption in SPM synthesis. SPMs hold great promise for neuroprotection in AD by altering expression of pro-inflammatory genes, modulating macrophage function, serving as a biomarker for AD status, and promoting resolution of neuroinflammation. In PD, data suggest SPM are able to cross the blood-brain barrier, inhibit microglial activation and decrease induced markers of inflammation, possibly as a result of their ability to downregulate NFκB signaling pathways. Several in vivo and in vitro studies suggest a benefit from administration of SPMs in both neurodegenerative disorders. However, extrapolation of these outcomes to humans is difficult as no models are able to replicate all features of AD or PD. Minimal data evaluating these PUFA-derived metabolites in humans with neurodegenerative disorders are available and a gap in knowledge exists regarding behavior of SPM and their receptors in patients with these conditions. There is also large gap in our knowledge regarding which lipid mediator would be most effective in which model of AD or PD and how dietary intake or supplementation can impact SPM levels. Future direction should include focused, translational efforts to investigate SPM as an add-on (in addition to standard treatment) or as standalone agents in patients with neurodegenerative disorders.
Collapse
Affiliation(s)
- Jana Ponce
- Division of Medical Nutrition Education, College of Allied Health Professions, University of Nebraska Medical Center, Omaha, NE, United States
- *Correspondence: Jana Ponce,
| | - Arzu Ulu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Corrine Hanson
- Division of Medical Nutrition Education, College of Allied Health Professions, University of Nebraska Medical Center, Omaha, NE, United States
| | - Erin Cameron-Smith
- Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - John Bertoni
- Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jenna Wuebker
- Department of Pharmaceutical and Nutrition Care, Nebraska Medicine, Omaha, NE, United States
| | - Alfred Fisher
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Ka-Chun Siu
- Division of Medical Nutrition Education, College of Allied Health Professions, University of Nebraska Medical Center, Omaha, NE, United States
| | - Vivien Marmelat
- Department of Biomechanics, College of Education, Health, and Human Sciences, University of Nebraska - Omaha, Omaha, NE, United States
| | - Jiri Adamec
- Department of Biochemistry, College of Arts and Sciences, University of Nebraska - Lincoln, Lincoln, NE, United States
| | - Danish Bhatti
- Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
10
|
Ordóñez-Gutiérrez L, Fábrias G, Casas J, Wandosell F. Diets with Higher ω-6/ω-3 Ratios Show Differences in Ceramides and Fatty Acid Levels Accompanied by Increased Amyloid-Beta in the Brains of Male APP/PS1 Transgenic Mice. Int J Mol Sci 2021; 22:ijms222010907. [PMID: 34681567 PMCID: PMC8535881 DOI: 10.3390/ijms222010907] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022] Open
Abstract
Senile plaque formation as a consequence of amyloid-β peptide (Aβ) aggregation constitutes one of the main hallmarks of Alzheimer's disease (AD). This pathology is characterized by synaptic alterations and cognitive impairment. In order to either prevent or revert it, different therapeutic approaches have been proposed, and some of them are focused on diet modification. Modification of the ω-6/ω-3 fatty acids (FA) ratio in diets has been proven to affect Aβ production and senile plaque formation in the hippocampus and cortex of female transgenic (TG) mice. In these diets, linoleic acid is the main contribution of ω-6 FA, whereas alpha-linoleic acid (ALA), eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA) and docosapentaenoic acid (DPA) are the contributors of ω-3 FA. In the present work, we have explored the effect of ω-6/ω-3 ratio modifications in the diets of male double-transgenic APPswe/PS1ΔE9 (AD model) and wild-type mice (WT). Amyloid burden in the hippocampus increased in parallel with the increase in dietary ω-6/ω-3 ratio in TG male mice. In addition, there was a modification in the brain lipid profile proportional to the ω-6/ω-3 ratio of the diet. In particular, the higher the ω-6/ω-3 ratio, the lower the ceramides and higher the FAs, particularly docosatetraenoic acid. Modifications to the cortex lipid profile was mostly similar between TG and WT mice, except for gangliosides (higher levels in TG mice) and some ceramide species (lower levels in TG mice).
Collapse
Affiliation(s)
- Lara Ordóñez-Gutiérrez
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain;
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 28049 Madrid, Spain
| | - Gemma Fábrias
- Instituto de Química Avanzada de Cataluña (IQAC-CSIC), 080034 Barcelona, Spain; (G.F.); (J.C.)
| | - Josefina Casas
- Instituto de Química Avanzada de Cataluña (IQAC-CSIC), 080034 Barcelona, Spain; (G.F.); (J.C.)
| | - Francisco Wandosell
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain;
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 28049 Madrid, Spain
- Correspondence: ; Tel.: +34-91-196-4561
| |
Collapse
|
11
|
Lessard-Beaudoin M, M Gonzalez L, AlOtaibi M, Chouinard-Watkins R, Plourde M, Calon F, Graham RK. Diet enriched in omega-3 fatty acids alleviates olfactory system deficits in APOE4 transgenic mice. Eur J Neurosci 2021; 54:7092-7108. [PMID: 34549475 DOI: 10.1111/ejn.15472] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 11/30/2022]
Abstract
Olfactory dysfunction is observed in several neurological disorders including Mild Cognitive Impairment (MCI) and Alzheimer disease (AD). These deficits occur early and correlate with global cognitive performance, depression and degeneration of olfactory regions in the brain. Despite extensive human studies, there has been little characterization of the olfactory system in models of AD. In order to determine if olfactory structural and/or molecular phenotypes are observed in a model expressing a genetic risk factor for AD, we assessed the olfactory bulb (OB) in APOE4 transgenic mice. A significant decrease in OB weight was observed at 12 months of age in APOE4 mice concurrent with inflammation and decreased NeuN expression. In order to determine if a diet rich in omega-3s may alleviate the olfactory system phenotypes observed, we assessed WT and APOE4 mice on a docosahexaenoic acid (DHA) diet. APOE4 mice on a DHA diet did not present with atrophy of the OB, and the alterations in NeuN and IBA-1 expression were alleviated. Furthermore, alterations in caspase mRNA and protein expression in the APOE4 OB were not observed with a DHA diet. Similar to the human AD condition, OB atrophy is an early phenotype in the APOE4 mice and concurrent with inflammation. These data support a link between the structural olfactory brain region atrophy and the olfactory dysfunction observed in AD and suggest that inflammation and cell death pathways may contribute to the olfactory deficits observed. Furthermore, the results suggest that diets enriched in DHA may provide benefit to APOE4 allele carriers.
Collapse
Affiliation(s)
- Melissa Lessard-Beaudoin
- Research Center on Aging, CIUSSS de L'Estrie - CHUS, Sherbrooke, Quebec, Canada.,Department of Pharmacology and Physiology, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Laura M Gonzalez
- Research Center on Aging, CIUSSS de L'Estrie - CHUS, Sherbrooke, Quebec, Canada.,Department of Pharmacology and Physiology, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Majed AlOtaibi
- Research Center on Aging, CIUSSS de L'Estrie - CHUS, Sherbrooke, Quebec, Canada.,Department of Pharmacology and Physiology, University of Sherbrooke, Sherbrooke, Quebec, Canada.,Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - Raphaël Chouinard-Watkins
- Research Center on Aging, CIUSSS de L'Estrie - CHUS, Sherbrooke, Quebec, Canada.,Department of Medicine, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Melanie Plourde
- Research Center on Aging, CIUSSS de L'Estrie - CHUS, Sherbrooke, Quebec, Canada.,Department of Medicine, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Frederic Calon
- Faculty of Pharmacy, Centre de Recherche du CHU de Québec, Université Laval, Quebec City, Quebec, Canada
| | - Rona K Graham
- Research Center on Aging, CIUSSS de L'Estrie - CHUS, Sherbrooke, Quebec, Canada.,Department of Pharmacology and Physiology, University of Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
12
|
Peng Y, Tao H, Wang S, Xiao J, Wang Y, Su H. Dietary intervention with edible medicinal plants and derived products for prevention of Alzheimer's disease: A compendium of time-tested strategy. J Funct Foods 2021; 81:104463. [DOI: 10.1016/j.jff.2021.104463] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
13
|
Baumel BS, Doraiswamy PM, Sabbagh M, Wurtman R. Potential Neuroregenerative and Neuroprotective Effects of Uridine/Choline-Enriched Multinutrient Dietary Intervention for Mild Cognitive Impairment: A Narrative Review. Neurol Ther 2021; 10:43-60. [PMID: 33368017 PMCID: PMC8139993 DOI: 10.1007/s40120-020-00227-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/02/2020] [Indexed: 01/21/2023] Open
Abstract
In mild cognitive impairment (MCI) due to Alzheimer disease (AD), also known as prodromal AD, there is evidence for a pathologic shortage of uridine, choline, and docosahexaenoic acid [DHA]), which are key nutrients needed by the brain. Preclinical and clinical evidence shows the importance of nutrient bioavailability to support the development and maintenance of brain structure and function in MCI and AD. Availability of key nutrients is limited in MCI, creating a distinct nutritional need for uridine, choline, and DHA. Evidence suggests that metabolic derangements associated with ageing and disease-related pathology can affect the body's ability to generate and utilize nutrients. This is reflected in lower levels of nutrients measured in the plasma and brains of individuals with MCI and AD dementia, and progressive loss of cognitive performance. The uridine shortage cannot be corrected by normal diet, making uridine a conditionally essential nutrient in affected individuals. It is also challenging to correct the choline shortfall through diet alone, because brain uptake from the plasma significantly decreases with ageing. There is no strong evidence to support the use of single-agent supplements in the management of MCI due to AD. As uridine and choline work synergistically with DHA to increase phosphatidylcholine formation, there is a compelling rationale to combine these nutrients. A multinutrient enriched with uridine, choline, and DHA developed to support brain function has been evaluated in randomized controlled trials covering a spectrum of dementia from MCI to moderate AD. A randomized controlled trial in subjects with prodromal AD showed that multinutrient intervention slowed brain atrophy and improved some measures of cognition. Based on the available clinical evidence, nutritional intervention should be considered as a part of the approach to the management of individuals with MCI due to AD, including adherence to a healthy, balanced diet, and consideration of evidence-based multinutrient supplements.
Collapse
Affiliation(s)
- Barry S Baumel
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - P Murali Doraiswamy
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Marwan Sabbagh
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Las Vegas, NV, USA
| | - Richard Wurtman
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
14
|
Fu CX, Dai L, Yuan XY, Xu YJ. Effects of Fish Oil Combined with Selenium and Zinc on Learning and Memory Impairment in Aging Mice and Amyloid Precursor Protein Processing. Biol Trace Elem Res 2021; 199:1855-1863. [PMID: 32666432 DOI: 10.1007/s12011-020-02280-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/06/2020] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease is characterized by the aggregation of amyloid-beta (Aβ) peptide into plaques and neurofibrillary tangles. Aβ peptide is generated by the cleavage of the β-amyloid precursor protein (APP) by β- and γ-secretase. The present study was conducted to investigate the effects of fish oil (or eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)), selenium, and zinc on learning and memory impairment in an aging mouse model and on APP. We performed the Morris water maze and platform recorder tests on male Kunming mice (10/group) grouped as control and D-galactose-induced aging model mice treated with vehicle, fish oil, fish oil + selenium, fish oil + selenium + zinc, and positive control (red ginseng extract). Fish oil + zinc + selenium for 7 weeks significantly improved learning and memory impairments in aging model animals in the Morris water maze and platform recorder tests, as evidenced by shortened incubation periods and number of errors. In vitro analysis of Aβ1-40 content in APP695-transfected CHO cells revealed a decrease after treatment with EPA, DHA, and their combinations with selenium or selenium and zinc. Assaying β- and γ-secretase activities revealed a decrease in PC12 cells and mouse serum as well as a decrease in β-site APP-cleaving enzyme 1 and presenilin 1 protein levels in the PC12 cells and mouse serum. Taken together, our results show that fish oil combined with selenium and zinc inhibited APP processing and alleviated learning and memory impairment in a mouse model of aging.
Collapse
Affiliation(s)
- Chao-Xu Fu
- Department of Preventive Medicine, Medical College of Yanbian University, Yanji, Jilin, China
| | - Lin Dai
- Department of Preventive Medicine, Medical College of Yanbian University, Yanji, Jilin, China
| | - Xiu-Yuan Yuan
- Department of Preventive Medicine, Medical College of Yanbian University, Yanji, Jilin, China
| | - Yan-Ji Xu
- Department of Preventive Medicine, Medical College of Yanbian University, Yanji, Jilin, China.
| |
Collapse
|
15
|
Martens N, Schepers M, Zhan N, Leijten F, Voortman G, Tiane A, Rombaut B, Poisquet J, Sande NVD, Kerksiek A, Kuipers F, Jonker JW, Liu H, Lütjohann D, Vanmierlo T, Mulder MT. 24(S)-Saringosterol Prevents Cognitive Decline in a Mouse Model for Alzheimer's Disease. Mar Drugs 2021; 19:190. [PMID: 33801706 PMCID: PMC8065937 DOI: 10.3390/md19040190] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/16/2021] [Accepted: 03/25/2021] [Indexed: 12/13/2022] Open
Abstract
We recently found that dietary supplementation with the seaweed Sargassum fusiforme, containing the preferential LXRβ-agonist 24(S)-saringosterol, prevented memory decline and reduced amyloid-β (Aβ) deposition in an Alzheimer's disease (AD) mouse model without inducing hepatic steatosis. Here, we examined the effects of 24(S)-saringosterol as a food additive on cognition and neuropathology in AD mice. Six-month-old male APPswePS1ΔE9 mice and wildtype C57BL/6J littermates received 24(S)-saringosterol (0.5 mg/25 g body weight/day) (APPswePS1ΔE9 n = 20; C57BL/6J n = 19) or vehicle (APPswePS1ΔE9 n = 17; C57BL/6J n = 19) for 10 weeks. Cognition was assessed using object recognition and object location tasks. Sterols were analyzed by gas chromatography/mass spectrometry, Aβ and inflammatory markers by immunohistochemistry, and gene expression by quantitative real-time PCR. Hepatic lipids were quantified after Oil-Red-O staining. Administration of 24(S)-saringosterol prevented cognitive decline in APPswePS1ΔE9 mice without affecting the Aβ plaque load. Moreover, 24(S)-saringosterol prevented the increase in the inflammatory marker Iba1 in the cortex of APPswePS1ΔE9 mice (p < 0.001). Furthermore, 24(S)-saringosterol did not affect the expression of lipid metabolism-related LXR-response genes in the hippocampus nor the hepatic neutral lipid content. Thus, administration of 24(S)-saringosterol prevented cognitive decline in APPswePS1ΔE9 mice independent of effects on Aβ load and without adverse effects on liver fat content. The anti-inflammatory effects of 24(S)-saringosterol may contribute to the prevention of cognitive decline.
Collapse
Affiliation(s)
- Nikita Martens
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, BE 3590 Hasselt, Belgium
| | - Melissa Schepers
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, BE 3590 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neurosciences, Division Translational Neuroscience, Maastricht University, 6200 Maastricht, The Netherlands
| | - Na Zhan
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266100, China
| | - Frank Leijten
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
| | - Gardi Voortman
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
| | - Assia Tiane
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, BE 3590 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neurosciences, Division Translational Neuroscience, Maastricht University, 6200 Maastricht, The Netherlands
| | - Ben Rombaut
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, BE 3590 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neurosciences, Division Translational Neuroscience, Maastricht University, 6200 Maastricht, The Netherlands
| | - Janne Poisquet
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, BE 3590 Hasselt, Belgium
| | - Nienke van de Sande
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neurosciences, Division Translational Neuroscience, Maastricht University, 6200 Maastricht, The Netherlands
| | - Anja Kerksiek
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53105 Bonn, Germany
| | - Folkert Kuipers
- Department of Pediatrics, Section of Molecular Metabolism and Nutrition, University Medical Center Groningen, 9713 Groningen, The Netherlands
| | - Johan W Jonker
- Department of Pediatrics, Section of Molecular Metabolism and Nutrition, University Medical Center Groningen, 9713 Groningen, The Netherlands
| | - Hongbing Liu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266100, China
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53105 Bonn, Germany
| | - Tim Vanmierlo
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, BE 3590 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neurosciences, Division Translational Neuroscience, Maastricht University, 6200 Maastricht, The Netherlands
| | - Monique T Mulder
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
| |
Collapse
|
16
|
van Oudenhoven FM, Swinkels SHN, Soininen H, Kivipelto M, Hartmann T, Rizopoulos D. A competing risk joint model for dealing with different types of missing data in an intervention trial in prodromal Alzheimer's disease. Alzheimers Res Ther 2021; 13:63. [PMID: 33752738 PMCID: PMC7983401 DOI: 10.1186/s13195-021-00801-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/04/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND Missing data can complicate the interpretability of a clinical trial, especially if the proportion is substantial and if there are different, potentially outcome-dependent causes. METHODS We aimed to obtain unbiased estimates, in the presence of a high level of missing data, for the intervention effects in a prodromal Alzheimer's disease trial: the LipiDiDiet study. We used a competing risk joint model that can simultaneously model each patient's longitudinal outcome trajectory in combination with the timing and type of missingness. RESULTS Using the competing risk joint model, we were able to provide unbiased estimates of the intervention effects in the presence of the different types of missingness. For the LipiDiDiet study, the intervention effects remained statistically significant after this correction for the timing and type of missingness. CONCLUSION Missing data is a common problem in (Alzheimer) clinical trials. It is important to realize that statistical techniques make specific assumptions about the missing data mechanisms. When there are different missing data sources, a competing risk joint model is a powerful method because it can explicitly model the association between the longitudinal data and each type of missingness. TRIAL REGISTRATION Dutch Trial Register, NTR1705 . Registered on 9 March 2009.
Collapse
Affiliation(s)
- Floor M van Oudenhoven
- Department of Biostatistics, Erasmus Medical Center, PO Box 2040, 3000, Rotterdam, CA, the Netherlands.
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT, Utrecht, The Netherlands.
| | | | - Hilkka Soininen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, PO Box 1627, 70211, Kuopio, Finland
- Neurocenter, Department of Neurology, Kuopio University Hospital, PO Box 100, 70029, Kuopio, Finland
| | - Miia Kivipelto
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, PO Box 1627, 70211, Kuopio, Finland
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, 14157, Huddinge, Sweden
- Clinical Trials Unit, Theme Aging, Karolinska University Hospital, 14152, Huddinge, Sweden
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, St Dunstan's Road, London, UK
| | - Tobias Hartmann
- Deutsches Institut für Demenz Prävention (DIDP), Medical Faculty, Saarland University, Kirrbergerstraße, 66421, Homburg, Germany
- Department of Experimental Neurology, Saarland University, Kirrbergerstraße, 66421, Homburg, Germany
| | - Dimitris Rizopoulos
- Department of Biostatistics, Erasmus Medical Center, PO Box 2040, 3000, Rotterdam, CA, the Netherlands
| |
Collapse
|
17
|
Jie F, Yang X, Wu L, Wang M, Lu B. Linking phytosterols and oxyphytosterols from food to brain health: origins, effects, and underlying mechanisms. Crit Rev Food Sci Nutr 2021; 62:3613-3630. [PMID: 33397124 DOI: 10.1080/10408398.2020.1867819] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Phytosterols and their oxidation products, namely oxyphytosterols, are natural compounds present in plant foods. With increased intake of phytosterol-enriched functional food products, the exposure of both phytosterols and oxyphytosterols is rising. Over the past ten years, researches have been focused on their absorption and metabolism in human body, as well as their biological effects. More importantly, recent studies showed that phytosterols and oxyphytosterols can traverse the blood-brain barrier and accumulate in the brain. As brain health problems resulting from ageing being more serious, attenuating central nervous system (CNS) disorders with active compounds in food are becoming a hot topic. Phytosterols and oxyphytosterols have been shown to implicated in cognition altering and the pathologies of several CNS disorders, including Alzheimer's disease and multiple sclerosis. We will overview these findings with a focus on the contents of phytosterols and oxyphytosterols in food and their dietary intake, as well as their origins in the brain, and illustrate molecular pathways through which they affect brain health, in terms of inflammation, cholesterol homeostasis, oxidative stress, and mitochondria function. The existing scientific gaps of phytosterols and oxyphytosterols to brain health in knowledge are also discussed, highlighting research directions in the future.
Collapse
Affiliation(s)
- Fan Jie
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, China.,Fuli Institute of Food Science, Zhejiang University, Hangzhou, China.,Ningbo Research Institute, Zhejiang University, Ningbo, China
| | - Xuan Yang
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
| | - Lipeng Wu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, China.,Fuli Institute of Food Science, Zhejiang University, Hangzhou, China.,Ningbo Research Institute, Zhejiang University, Ningbo, China
| | - Mengmeng Wang
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, China.,Fuli Institute of Food Science, Zhejiang University, Hangzhou, China.,Ningbo Research Institute, Zhejiang University, Ningbo, China
| | - Baiyi Lu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, China.,Fuli Institute of Food Science, Zhejiang University, Hangzhou, China.,Ningbo Research Institute, Zhejiang University, Ningbo, China
| |
Collapse
|
18
|
Corrigendum: Edible seaweed-derived constituents: an undisclosed source of neuroprotective compounds. Neural Regen Res 2021; 16:2564-2568. [PMID: 33907050 PMCID: PMC8374581 DOI: 10.4103/1673-5374.313070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
|
19
|
Soininen H, Solomon A, Visser PJ, Hendrix SB, Blennow K, Kivipelto M, Hartmann T. 36-month LipiDiDiet multinutrient clinical trial in prodromal Alzheimer's disease. Alzheimers Dement 2020; 17:29-40. [PMID: 32920957 PMCID: PMC7821311 DOI: 10.1002/alz.12172] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 07/10/2020] [Accepted: 07/14/2020] [Indexed: 12/28/2022]
Abstract
Introduction The LipiDiDiet trial investigates the effects of the specific multinutrient combination Fortasyn Connect on cognition and related measures in prodromal Alzheimer's disease (AD). Based on previous results we hypothesized that benefits increase with long‐term intervention. Methods In this randomized, double‐blind, placebo‐controlled trial, 311 people with prodromal AD were recruited using the International Working Group‐1 criteria and assigned to active product (125 mL once‐a‐day drink) or an isocaloric, same tasting, placebo control drink. Main outcome was change in cognition (Neuropsychological Test Battery [NTB] 5‐item composite). Analyses were by modified intention‐to‐treat, excluding (ie, censoring) data collected after the start of open‐label active product and/or AD medication. Results Of the 382 assessed for eligibility, 311 were randomized, of those 162 participants completed the 36‐month study, including 81 with 36‐month data eligible for efficacy analysis. Over 36 months, significant reductions in decline were observed for the NTB 5‐item composite (−60%; between‐group difference 0.212 [95% confidence interval: 0.044 to 0.380]; P = 0.014), Clinical Dementia Rating‐Sum of Boxes (−45%; P = 0.014), memory (−76%; P = 0.008), and brain atrophy measures; small to medium Cohen's d effect size (0.25–0.31) similar to established clinically relevant AD treatment. Discussion This multinutrient intervention slowed decline on clinical and other measures related to cognition, function, brain atrophy, and disease progression. These results indicate that intervention benefits increased with long‐term use.
Collapse
Affiliation(s)
- Hilkka Soininen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland.,Neurocenter, Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Alina Solomon
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland.,Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Huddinge, Sweden.,Clinical Trials Unit, Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Pieter Jelle Visser
- Department of Neurology, Alzheimer Center, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands.,Department of Psychiatry and Neuropsychology, Alzheimer Center Limburg, University of Maastricht, Maastricht, the Netherlands
| | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Miia Kivipelto
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland.,Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Huddinge, Sweden.,Clinical Trials Unit, Theme Aging, Karolinska University Hospital, Huddinge, Sweden.,Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland.,Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London, United Kingdom
| | - Tobias Hartmann
- Deutsches Institut für Demenz Prävention (DIDP), Medical Faculty, Saarland University, Kirrbergerstraße, Homburg, Germany.,Department of Experimental Neurology, Saarland University, Kirrbergerstraße, Homburg, Germany
| | | |
Collapse
|
20
|
Dierckx T, Bogie JFJ, Hendriks JJA. The Impact of Phytosterols on the Healthy and Diseased Brain. Curr Med Chem 2020; 26:6750-6765. [PMID: 29984647 DOI: 10.2174/0929867325666180706113844] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/28/2018] [Accepted: 03/07/2018] [Indexed: 02/07/2023]
Abstract
The central nervous system (CNS) is the most cholesterol-rich organ in mammals. Cholesterol homeostasis is essential for proper brain functioning and dysregulation of cholesterol metabolism can lead to neurological problems. Multiple sclerosis (MS) and Alzheimer's disease (AD) are examples of neurological diseases that are characterized by a disturbed cholesterol metabolism. Phytosterols (PS) are plant-derived components that structurally and functionally resemble cholesterol. PS are known for their cholesterol-lowering properties. Due to their ability to reach the brain, researchers have started to investigate the physiological role of PS in the CNS. In this review, the metabolism and function of PS in the diseased and healthy CNS are discussed.
Collapse
Affiliation(s)
- Tess Dierckx
- Biomedical Research Institute, Hasselt University, Diepenbeek, Hassett, Belgium
| | - Jeroen F J Bogie
- Biomedical Research Institute, Hasselt University, Diepenbeek, Hassett, Belgium
| | - Jerome J A Hendriks
- Biomedical Research Institute, Hasselt University, Diepenbeek, Hassett, Belgium
| |
Collapse
|
21
|
Schepers M, Martens N, Tiane A, Vanbrabant K, Liu HB, Lütjohann D, Mulder M, Vanmierlo T. Edible seaweed-derived constituents: an undisclosed source of neuroprotective compounds. Neural Regen Res 2020; 15:790-795. [PMID: 31719238 PMCID: PMC6990778 DOI: 10.4103/1673-5374.268894] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Edible marine algae, or seaweeds, are a rich source of several bioactive compounds including phytosterols, carotenoids, and polysaccharides. Over the last decades, seaweed-derived constituents turned out to not only reside in the systemic circulation, but are able to cross the blood-brain barrier to exert neuro-active functions both in homeostatic and pathological conditions. Therefore, seaweed-derived constituents have gained increasing interest for their neuro-immunomodulatory and neuroprotective properties, rendering them interesting candidates for the management of several neurodegenerative disorders. In particular seaweed-derived phytosterols gained interest for the treatment of neurodegenerative disorders as they potentiate neuroplasticity, enhance phagocytic clearance of neurotoxic peptides and have anti-inflammatory properties. Though, the anti-inflammatory and anti-oxidative properties of other constituents including carotenoids, phenols and polysaccharides have recently gained more interest. In this review, we provide an overview of a selection of the described neuro-active properties of seaweed-derived constituents with a focus on phytosterols.
Collapse
Affiliation(s)
- Melissa Schepers
- Department of Neuroimmunology, Biomedical Research Institute, Hasselt University, European Graduate School of Neuroscience (EURON), Hasselt, Belgium; Department of Psychiatry & Neuropsychology, Division of Translational Neuroscience, School for Mental Health and Neuroscience, Maastricht University, European Graduate School of Neuroscience (EURON), Maastricht, The Netherlands
| | - Nikita Martens
- Department of Internal Medicine, Laboratory of Vascular Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Assia Tiane
- Department of Neuroimmunology, Biomedical Research Institute, Hasselt University, European Graduate School of Neuroscience (EURON), Hasselt, Belgium; Department of Psychiatry & Neuropsychology, Division of Translational Neuroscience, School for Mental Health and Neuroscience, Maastricht University, European Graduate School of Neuroscience (EURON), Maastricht, The Netherlands
| | - Kenneth Vanbrabant
- Department of Neuroimmunology, Biomedical Research Institute, Hasselt University, European Graduate School of Neuroscience (EURON), Hasselt, Belgium; Institue for Clinical Chemistry and Clinical Pharmacology, Bonn, Germany
| | - Hong-Bing Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong Province, China
| | - Dieter Lütjohann
- Institue for Clinical Chemistry and Clinical Pharmacology, Bonn, Germany
| | - Monique Mulder
- Department of Internal Medicine, Laboratory of Vascular Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Tim Vanmierlo
- Department of Neuroimmunology, Biomedical Research Institute, Hasselt University, European Graduate School of Neuroscience (EURON), Hasselt, Belgium; Department of Psychiatry & Neuropsychology, Division of Translational Neuroscience, School for Mental Health and Neuroscience, Maastricht University, European Graduate School of Neuroscience (EURON), Maastricht, The Netherlands
| |
Collapse
|
22
|
Rasmussen J. The LipiDiDiet trial: what does it add to the current evidence for Fortasyn Connect in early Alzheimer's disease? Clin Interv Aging 2019; 14:1481-1492. [PMID: 31616139 PMCID: PMC6699494 DOI: 10.2147/cia.s211739] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 06/14/2019] [Indexed: 11/23/2022] Open
Abstract
Nutritional factors can influence the risk of developing Alzheimer’s disease (AD) and its rate of progression, and there is, therefore, increasing interest in nutrition as a modifiable risk factor for the disease. Synaptic loss is an important feature of early AD, and the formation of new synapses is dependent on key nutritional elements that are known to be deficient in patients with AD. The daily medical food, Souvenaid, contains Fortasyn Connect, a multinutrient combination developed to specifically address these deficiencies, comprising docosahexaenoic acid, eicosapentaenoic acid, uridine monophosphate, choline, phospholipids, selenium, folic acid, and vitamins B12, B6, C, and E. Although yielding heterogeneous findings, clinical studies of Fortasyn Connect provide preliminary evidence of clinically relevant benefits on cognitive outcomes in prodromal and early AD. The LipiDiDiet trial investigated the effects of Fortasyn Connect on cognition and related measures in prodromal AD, and is the first randomized, controlled, double-blind, multicenter trial study of a non-pharmacological intervention in this setting. The primary efficacy endpoint was change over 24 months in a composite score of cognitive performance using a neuropsychological test battery. Fortasyn Connect had no significant effect on this endpoint, but demonstrated a significant benefit on secondary endpoints, including domains of cognition affected by AD (attention, memory, executive function) and hippocampal atrophy, suggesting a potential benefit on disease progression. Other studies have demonstrated benefits for Fortasyn Connect on nutritional markers and levels of plasma homocysteine. Taken together, current evidence indicates that Fortasyn Connect may show benefit on domains of cognition affected by AD and nutritional measures that influence risk factors for its progression; that it has greater potential for benefit earlier rather than later in the disease; and that it is safe and well tolerated, alone or in combination with AD medications. Further research into its potential role in AD management is therefore warranted.
Collapse
Affiliation(s)
- Jill Rasmussen
- Primary Care Specialist Mental Health in Dementia and Learning Disability, Surrey, UK.,Royal College of General Practitioners Representative for Dementia, London, UK
| |
Collapse
|
23
|
Dietary Sargassum fusiforme improves memory and reduces amyloid plaque load in an Alzheimer's disease mouse model. Sci Rep 2019; 9:4908. [PMID: 30894635 PMCID: PMC6426980 DOI: 10.1038/s41598-019-41399-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 03/05/2019] [Indexed: 12/12/2022] Open
Abstract
Activation of liver X receptors (LXRs) by synthetic agonists was found to improve cognition in Alzheimer's disease (AD) mice. However, these LXR agonists induce hypertriglyceridemia and hepatic steatosis, hampering their use in the clinic. We hypothesized that phytosterols as LXR agonists enhance cognition in AD without affecting plasma and hepatic triglycerides. Phytosterols previously reported to activate LXRs were tested in a luciferase-based LXR reporter assay. Using this assay, we found that phytosterols commonly present in a Western type diet in physiological concentrations do not activate LXRs. However, a lipid extract of the 24(S)-Saringosterol-containing seaweed Sargassum fusiforme did potently activate LXRβ. Dietary supplementation of crude Sargassum fusiforme or a Sargassum fusiforme-derived lipid extract to AD mice significantly improved short-term memory and reduced hippocampal Aβ plaque load by 81%. Notably, none of the side effects typically induced by full synthetic LXR agonists were observed. In contrast, administration of the synthetic LXRα activator, AZ876, did not improve cognition and resulted in the accumulation of lipid droplets in the liver. Administration of Sargassum fusiforme-derived 24(S)-Saringosterol to cultured neurons reduced the secretion of Aβ42. Moreover, conditioned medium from 24(S)-Saringosterol-treated astrocytes added to microglia increased phagocytosis of Aβ. Our data show that Sargassum fusiforme improves cognition and alleviates AD pathology. This may be explained at least partly by 24(S)-Saringosterol-mediated LXRβ activation.
Collapse
|
24
|
Milanovic D, Petrovic S, Brkic M, Avramovic V, Perovic M, Ivkovic S, Glibetic M, Kanazir S. Short-Term Fish Oil Treatment Changes the Composition of Phospholipids While Not Affecting the Expression of Mfsd2a Omega-3 Transporter in the Brain and Liver of the 5xFAD Mouse Model of Alzheimer's Disease. Nutrients 2018; 10:nu10091250. [PMID: 30200627 PMCID: PMC6165196 DOI: 10.3390/nu10091250] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/25/2018] [Accepted: 08/28/2018] [Indexed: 01/22/2023] Open
Abstract
Long-term fish oil (FO) supplementation is able to improve Alzheimer’s disease (AD) pathology. We aimed to determine the impact of short-term fish oil (FO) intake on phospholipids composition and plaque pathology in 5xFAD mice, a widely used animal model of AD. A 3-week-long FO supplementation administered at 3 months of age decreased the number of dense core plaques in the 5xFAD cortex and changed phospholipids in the livers and brains of wild-type (Wt) and 5xFAD mice. Livers of both genotypes responded by increase of n-3 and reciprocal decrease of n-6 fatty acids. In Wt brains, FO supplementation induced elevation of n-3 fatty acids and subsequent enhancement of n-6/n-3 ratio. However, in 5xFAD brains the improved n-6/n-3 ratio was mainly due to FO-induced decrease in arachidonic and adrenic n-6 fatty acids. Also, brain and liver abundance of n-3 fatty acids were strongly correlated in Wts, oppositely to 5xFADs where significant brain-liver correlation exists only for n-6 fatty acids. Expression of omega-3 transporter Mfs2a remained unchanged after FO supplementation. We have demonstrated that even a short-term FO intake improves the phospholipid composition and has a significant effect on plaque burden in 5xFAD brains when applied in early stages of AD pathology.
Collapse
Affiliation(s)
- Desanka Milanovic
- Department of Neurobiology, Institute for Biological Research, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia.
| | - Snjezana Petrovic
- Center of Excellence in Nutrition and Metabolism Research, CENM, Tadeusa Koscuska 1, 11000 Belgrade, Serbia.
| | - Marjana Brkic
- Department of Neurobiology, Institute for Biological Research, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia.
| | - Vladimir Avramovic
- Department of Neurobiology, Institute for Biological Research, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia.
| | - Milka Perovic
- Department of Neurobiology, Institute for Biological Research, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia.
| | - Sanja Ivkovic
- Department of Neurobiology, Institute for Biological Research, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia.
| | - Marija Glibetic
- Center of Excellence in Nutrition and Metabolism Research, CENM, Tadeusa Koscuska 1, 11000 Belgrade, Serbia.
| | - Selma Kanazir
- Department of Neurobiology, Institute for Biological Research, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia.
| |
Collapse
|
25
|
Reversal of memory and neuropsychiatric symptoms and reduced tau pathology by selenium in 3xTg-AD mice. Sci Rep 2018; 8:6431. [PMID: 29691439 PMCID: PMC5915484 DOI: 10.1038/s41598-018-24741-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 04/05/2018] [Indexed: 12/03/2022] Open
Abstract
Accumulation of amyloid-β plaques and tau contribute to the pathogenesis of Alzheimer’s disease (AD), but it is unclear whether targeting tau pathology by antioxidants independently of amyloid-β causes beneficial effects on memory and neuropsychiatric symptoms. Selenium, an essential antioxidant element reduced in the aging brain, prevents development of neuropathology in AD transgenic mice at early disease stages. The therapeutic potential of selenium for ameliorating or reversing neuropsychiatric and cognitive behavioral symptoms at late AD stages is largely unknown. Here, we evaluated the effects of chronic dietary sodium selenate supplementation for 4 months in female 3xTg-AD mice at 12–14 months of age. Chronic sodium selenate treatment efficiently reversed hippocampal-dependent learning and memory impairments, and behavior- and neuropsychiatric-like symptoms in old female 3xTg-AD mice. Selenium significantly decreased the number of aggregated tau-positive neurons and astrogliosis, without globally affecting amyloid plaques, in the hippocampus of 3xTg-AD mice. These results indicate that selenium treatment reverses AD-like memory and neuropsychiatric symptoms by a mechanism involving reduction of aggregated tau and/or reactive astrocytes but not amyloid pathology. These results suggest that sodium selenate could be part of a combined therapeutic approach for the treatment of memory and neuropsychiatric symptoms in advanced AD stages.
Collapse
|
26
|
Fiandaca MS, Mapstone M, Connors E, Jacobson M, Monuki ES, Malik S, Macciardi F, Federoff HJ. Systems healthcare: a holistic paradigm for tomorrow. BMC SYSTEMS BIOLOGY 2017; 11:142. [PMID: 29258513 PMCID: PMC5738174 DOI: 10.1186/s12918-017-0521-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 12/01/2017] [Indexed: 12/13/2022]
Abstract
Systems healthcare is a holistic approach to health premised on systems biology and medicine. The approach integrates data from molecules, cells, organs, the individual, families, communities, and the natural and man-made environment. Both extrinsic and intrinsic influences constantly challenge the biological networks associated with wellness. Such influences may dysregulate networks and allow pathobiology to evolve, resulting in early clinical presentation that requires astute assessment and timely intervention for successful mitigation. Herein, we describe the components of relevant biological systems and the nature of progression from at-risk to manifest disease. We illustrate the systems approach by examining two relevant clinical examples: Alzheimer's and cardiovascular diseases. The implications of systems healthcare management are examined through the lens of economics, ethics, policy and the law. Finally, we propose the need to develop new educational paradigms to enhance the training of the health professional in an era of systems medicine.
Collapse
Affiliation(s)
- Massimo S Fiandaca
- Department of Neurology, School of Medicine, Irvine, USA
- Department of Neurological Surgery, School of Medicine, Irvine, USA
- Department of Anatomy & Neurobiology, School of Medicine, Irvine, USA
| | - Mark Mapstone
- Department of Neurology, School of Medicine, Irvine, USA
| | | | - Mireille Jacobson
- Department of Economics, Paul Merage School of Business, Irvine, USA
| | - Edwin S Monuki
- Department of Pathology & Laboratory Medicine, School of Medicine, Irvine, USA
| | - Shaista Malik
- Department of Medicine, School of Medicine, Irvine, USA
| | - Fabio Macciardi
- Department of Psychiatry & Human Behavior, School of Medicine, Irvine, USA
| | - Howard J Federoff
- Department of Neurology, School of Medicine, Irvine, USA.
- University of California Irvine (UCI), Irvine, CA, USA.
| |
Collapse
|
27
|
Grimm MOW, Michaelson DM, Hartmann T. Omega-3 fatty acids, lipids, and apoE lipidation in Alzheimer's disease: a rationale for multi-nutrient dementia prevention. J Lipid Res 2017; 58:2083-2101. [PMID: 28528321 PMCID: PMC5665674 DOI: 10.1194/jlr.r076331] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/09/2017] [Indexed: 12/14/2022] Open
Abstract
In the last decade, it has become obvious that Alzheimer's disease (AD) is closely linked to changes in lipids or lipid metabolism. One of the main pathological hallmarks of AD is amyloid-β (Aβ) deposition. Aβ is derived from sequential proteolytic processing of the amyloid precursor protein (APP). Interestingly, both, the APP and all APP secretases are transmembrane proteins that cleave APP close to and in the lipid bilayer. Moreover, apoE4 has been identified as the most prevalent genetic risk factor for AD. ApoE is the main lipoprotein in the brain, which has an abundant role in the transport of lipids and brain lipid metabolism. Several lipidomic approaches revealed changes in the lipid levels of cerebrospinal fluid or in post mortem AD brains. Here, we review the impact of apoE and lipids in AD, focusing on the major brain lipid classes, sphingomyelin, plasmalogens, gangliosides, sulfatides, DHA, and EPA, as well as on lipid signaling molecules, like ceramide and sphingosine-1-phosphate. As nutritional approaches showed limited beneficial effects in clinical studies, the opportunities of combining different supplements in multi-nutritional approaches are discussed and summarized.
Collapse
Affiliation(s)
- Marcus O W Grimm
- Department of Experimental Neurology and Department of Neurodegeneration and Neurobiology, and Deutsches Institut für DemenzPrävention (DIDP), Saarland University, Homburg/Saar, Germany
| | - Daniel M Michaelson
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Tobias Hartmann
- Department of Experimental Neurology and Department of Neurodegeneration and Neurobiology, and Deutsches Institut für DemenzPrävention (DIDP), Saarland University, Homburg/Saar, Germany
| |
Collapse
|
28
|
Soininen H, Solomon A, Visser PJ, Hendrix SB, Blennow K, Kivipelto M, Hartmann T. 24-month intervention with a specific multinutrient in people with prodromal Alzheimer's disease (LipiDiDiet): a randomised, double-blind, controlled trial. Lancet Neurol 2017; 16:965-975. [PMID: 29097166 PMCID: PMC5697936 DOI: 10.1016/s1474-4422(17)30332-0] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 09/06/2017] [Accepted: 09/12/2017] [Indexed: 01/31/2023]
Abstract
Background Nutrition is an important modifiable risk factor in Alzheimer's disease. Previous trials of the multinutrient Fortasyn Connect showed benefits in mild Alzheimer's disease dementia. LipiDiDiet investigated the effects of Fortasyn Connect on cognition and related measures in prodromal Alzheimer's disease. Here, we report the 24-month results of the trial. Methods LipiDiDiet was a 24-month randomised, controlled, double-blind, parallel-group, multicentre trial (11 sites in Finland, Germany, the Netherlands, and Sweden), with optional 12-month double-blind extensions. The trial enrolled individuals with prodromal Alzheimer's disease, defined according to the International Working Group (IWG)-1 criteria. Participants were randomly assigned (1:1) to active product (125 mL once-a-day drink containing Fortasyn Connect) or control product. Randomisation was computer-generated centrally in blocks of four, stratified by site. All study personnel and participants were masked to treatment assignment. The primary endpoint was change in a neuropsychological test battery (NTB) score. Analysis was by modified intention to treat. Safety analyses included all participants who consumed at least one study product dose. This trial is registered with the Dutch Trial Register, number NTR1705. Findings Between April 20, 2009, and July 3, 2013, 311 of 382 participants screened were randomly assigned to the active group (n=153) or control group (n=158). Mean change in NTB primary endpoint was −0·028 (SD 0·453) in the active group and −0·108 (0·528) in the control group; estimated mean treatment difference was 0·098 (95% CI −0·041 to 0·237; p=0·166). The decline in the control group was less than the prestudy estimate of −0·4 during 24 months. 66 (21%) participants dropped out of the study. Serious adverse events occurred in 34 (22%) participants in the active group and 30 (19%) in control group (p=0·487), none of which were regarded as related to the study intervention. Interpretation The intervention had no significant effect on the NTB primary endpoint over 2 years in prodromal Alzheimer's disease. However, cognitive decline in this population was much lower than expected, rendering the primary endpoint inadequately powered. Group differences on secondary endpoints of disease progression measuring cognition and function and hippocampal atrophy were observed. Further study of nutritional approaches with larger sample sizes, longer duration, or a primary endpoint more sensitive in this pre-dementia population, is needed. Funding European Commission 7th Framework Programme.
Collapse
Affiliation(s)
- Hilkka Soininen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland; Neurocenter, Department of Neurology, Kuopio University Hospital, Kuopio, Finland.
| | - Alina Solomon
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland; Department of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Huddinge, Sweden; Clinical Trials Unit, Department of Geriatric Medicine, Karolinska University Hospital, Huddinge, Sweden
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, Alzheimer Center Limburg, University of Maastricht, Maastricht, Netherlands; Department of Neurology, Alzheimer Center, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, Netherlands
| | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Miia Kivipelto
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland; Department of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Huddinge, Sweden; Clinical Trials Unit, Department of Geriatric Medicine, Karolinska University Hospital, Huddinge, Sweden
| | - Tobias Hartmann
- German Institute for Dementia Prevention (DIDP), Medical Faculty, and Department of Experimental Neurology, Saarland University, Homburg, Germany
| | | |
Collapse
|
29
|
Scheltens NME, Kuyper IS, Boellaard R, Barkhof F, Teunissen CE, Broersen LM, Lansbergen MM, van der Flier WM, van Berckel BNM, Scheltens P. Design of the NL-ENIGMA study: Exploring the effect of Souvenaid on cerebral glucose metabolism in early Alzheimer's disease. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2017; 2:233-240. [PMID: 29067310 PMCID: PMC5651347 DOI: 10.1016/j.trci.2016.07.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Introduction Alzheimer's disease is associated with early synaptic loss. Specific nutrients are known to be rate limiting for synapse formation. Studies have shown that administering specific nutrients may improve memory function, possibly by increasing synapse formation. This Dutch study explores the Effect of a specific Nutritional Intervention on cerebral Glucose Metabolism in early Alzheimer's disease (NL-ENIGMA, Dutch Trial Register NTR4718, http://www.trialregister.nl/trialreg/admin/rctview.asp?TC=4718). The NL-ENIGMA study is designed to test whether the specific multinutrient combination Fortasyn Connect present in the medical food Souvenaid influences cerebral glucose metabolism as a marker for improved synapse function. Methods This study is a double-blind, randomized controlled parallel-group single-center trial. Forty drug-naive patients with mild cognitive impairment or mild dementia with evidence of amyloid deposition are 1:1 randomized to receive either the multinutrient combination or placebo once daily. Main exploratory outcome parameters include absolute quantitative positron emission tomography with 18F-fluorodeoxyglucose (including arterial sampling) and standard uptake value ratios normalized for the cerebellum or pons after 24 weeks. Discussion We expect the NL-ENIGMA study to provide further insight in the potential of this multinutrient combination to improve synapse function. This study explores the Effect of a specific Nutritional Intervention on cerebral Glucose Metabolism in early Alzheimer's disease (AD). Forty drug-naive patients with mild cognitive impairment or mild dementia because of AD will be analyzed. Synapse function is explored using positron emission tomography with 18F-fluorodeoxyglucose including arterial sampling.
Collapse
Affiliation(s)
- Nienke M E Scheltens
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Ingrid S Kuyper
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands.,Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands.,Institutes of Neurology and Healthcare Engineering, UCL, London, UK
| | - Charlotte E Teunissen
- Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Laus M Broersen
- Nutricia Research, Nutricia Advanced Medical Nutrition, Utrecht, The Netherlands
| | - Marieke M Lansbergen
- Nutricia Research, Nutricia Advanced Medical Nutrition, Utrecht, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands.,Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | - Bart N M van Berckel
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Philip Scheltens
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Grimm MOW, Mett J, Grimm HS, Hartmann T. APP Function and Lipids: A Bidirectional Link. Front Mol Neurosci 2017; 10:63. [PMID: 28344547 PMCID: PMC5344993 DOI: 10.3389/fnmol.2017.00063] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 02/24/2017] [Indexed: 12/14/2022] Open
Abstract
Extracellular neuritic plaques, composed of aggregated amyloid-β (Aβ) peptides, are one of the major histopathological hallmarks of Alzheimer's disease (AD), a progressive, irreversible neurodegenerative disorder and the most common cause of dementia in the elderly. One of the most prominent risk factor for sporadic AD, carrying one or two aberrant copies of the apolipoprotein E (ApoE) ε4 alleles, closely links AD to lipids. Further, several lipid classes and fatty acids have been reported to be changed in the brain of AD-affected individuals. Interestingly, the observed lipid changes in the brain seem not only to be a consequence of the disease but also modulate Aβ generation. In line with these observations, protective lipids being able to decrease Aβ generation and also potential negative lipids in respect to AD were identified. Mechanistically, Aβ peptides are generated by sequential proteolytic processing of the amyloid precursor protein (APP) by β- and γ-secretase. The α-secretase appears to compete with β-secretase for the initial cleavage of APP, preventing Aβ production. All APP-cleaving secretases as well as APP are transmembrane proteins, further illustrating the impact of lipids on Aβ generation. Beside the pathological impact of Aβ, accumulating evidence suggests that Aβ and the APP intracellular domain (AICD) play an important role in regulating lipid homeostasis, either by direct effects or by affecting gene expression or protein stability of enzymes involved in the de novo synthesis of different lipid classes. This review summarizes the current literature addressing the complex bidirectional link between lipids and AD and APP processing including lipid alterations found in AD post mortem brains, lipids that alter APP processing and the physiological functions of Aβ and AICD in the regulation of several lipid metabolism pathways.
Collapse
Affiliation(s)
- Marcus O. W. Grimm
- Experimental Neurology, Saarland UniversityHomburg/Saar, Germany
- Neurodegeneration and Neurobiology, Saarland UniversityHomburg/Saar, Germany
- Deutsches Institut für DemenzPrävention (DIDP), Saarland UniversityHomburg/Saar, Germany
| | - Janine Mett
- Experimental Neurology, Saarland UniversityHomburg/Saar, Germany
| | - Heike S. Grimm
- Experimental Neurology, Saarland UniversityHomburg/Saar, Germany
| | - Tobias Hartmann
- Experimental Neurology, Saarland UniversityHomburg/Saar, Germany
- Neurodegeneration and Neurobiology, Saarland UniversityHomburg/Saar, Germany
- Deutsches Institut für DemenzPrävention (DIDP), Saarland UniversityHomburg/Saar, Germany
| |
Collapse
|
31
|
Reversal of high fat diet-induced obesity improves glucose tolerance, inflammatory response, β-amyloid accumulation and cognitive decline in the APP/PSEN1 mouse model of Alzheimer's disease. Neurobiol Dis 2017; 100:87-98. [PMID: 28108292 DOI: 10.1016/j.nbd.2017.01.004] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 12/14/2016] [Accepted: 01/16/2017] [Indexed: 02/06/2023] Open
Abstract
This study assessed the extent to which high fat diet (HFD)-induced β-amyloid accumulation and cognitive decline in APP/PSEN1 mice are reversible through control of fat intake. Ten months of HFD (60% calories from fat) led to significant deficits in a 2-trial Y maze task, and nest building assay, and decreased voluntary locomotor activity. The HFD induced an inflammatory response, indicated by increased expression of several inflammatory markers. Substituting a low fat diet led to pronounced weight loss and correction of glucose intolerance, decreases in the inflammatory response, and improved performance on behavioral tasks in both wild-type and APP/PSEN1 transgenic mice. Insoluble β-amyloid levels, and extent of tau phosphorylation were also lower following dietary reversal in APP/PSEN1 mice compared to high fat-fed animals, indicating that the inflammatory response may have contributed to key pathogenic pathways in the Alzheimer's disease model. The data suggest that weight loss can be a vital strategy for cognitive protection, but also highlight potential mechanisms for intervention when sustained weight loss is not possible.
Collapse
|
32
|
Shuang R, Rui X, Wenfang L. Phytosterols and Dementia. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2016; 71:347-354. [PMID: 27663717 DOI: 10.1007/s11130-016-0574-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
As the aging of the world's population is becoming increasingly serious, dementia-related diseases have become a hot topic in public health research. In recent years, human epidemiological studies have focused on lipid metabolism disorders and dementia. The efficacy of phytosterol intake as a cholesterol-lowering agent has been demonstrated. Phytosterols directly serve as ligands of the nuclear receptors, peroxisome proliferator-activated receptors (PPARs), activating Sirtuin 1 (SIRT-1), which are involved in the regulation of lipid metabolism and the pathogenesis of dementia. Moreover, phytosterols mediate cell and membrane cholesterol efflux or beta amyloid (Aβ) metabolism, which have preventative and therapeutic effects on dementia. Additionally, incorporation of plant sterols in lipid rafts can effectively reduce dietary fat and alter the dietary composition of fiber, fat and cholesterol to regulate appetite and calories. Overall, the objectives of this review are to explore whether phytosterols are a potentially effective target for the prevention of dementia and to discuss a possible molecular mechanism by which phytosterols play a role in the pathogenesis of dementia via the PPARs-SIRT-1 pathway.
Collapse
Affiliation(s)
- Rong Shuang
- Department of Public Health School, Wuhan University of Science & Technology, Wuhan, 430065, China.
| | - Xu Rui
- Department of Public Health School, Wuhan University of Science & Technology, Wuhan, 430065, China
| | - Li Wenfang
- Department of Public Health School, Wuhan University of Science & Technology, Wuhan, 430065, China.
| |
Collapse
|
33
|
van Straaten ECW, de Waal H, Lansbergen MM, Scheltens P, Maestu F, Nowak R, Hillebrand A, Stam CJ. Magnetoencephalography for the Detection of Intervention Effects of a Specific Nutrient Combination in Patients with Mild Alzheimer's Disease: Results from an Exploratory Double-Blind, Randomized, Controlled Study. Front Neurol 2016; 7:161. [PMID: 27799918 PMCID: PMC5065957 DOI: 10.3389/fneur.2016.00161] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/13/2016] [Indexed: 01/01/2023] Open
Abstract
Synaptic loss is an early pathological finding in Alzheimer’s disease (AD) and correlates with memory impairment. Changes in macroscopic brain activity measured with electro- and magnetoencephalography (EEG and MEG) in AD indicate synaptic changes and may therefore serve as markers of intervention effects in clinical trials. EEG peak frequency and functional networks have shown, in addition to improved memory performance, to be sensitive to detect an intervention effect in mild AD patients of the medical food Souvenaid containing the specific nutrient combination Fortasyn® Connect, which is designed to enhance synapse formation and function. Here, we explore the value of MEG, with higher spatial resolution than EEG, in identifying intervention effects of the nutrient combination by comparing MEG spectral measures, functional connectivity, and networks between an intervention and a control group. Quantitative markers describing spectral properties, functional connectivity, and graph theoretical aspects of MEG from the exploratory 24-week, double-blind, randomized, controlled Souvenir II MEG sub-study (NTR1975, http://www.trialregister.nl) in drug naïve patients with mild AD were compared between a test group (n = 27), receiving Souvenaid, and a control group (n = 28), receiving an isocaloric control product. The groups were unbalanced at screening with respect to Mini-Mental State Examination. Peak frequencies of MEG were compared with EEG peak frequencies, recorded in the same patients at similar time points, were compared with respect to sensitivity to intervention effects. No consistent statistically significant intervention effects were detected. In addition, we found no difference in sensitivity between MEG and EEG peak frequency. This exploratory study could not unequivocally establish the value of MEG in detecting interventional effects on brain activity, possibly due to small sample size and unbalanced study groups. We found no indication that the difference could be attributed to a lack of sensitivity of MEG compared with EEG. MEG in randomized controlled trials is feasible but its value to disclose intervention effects of Souvenaid in mild AD patients needs to be studied further.
Collapse
Affiliation(s)
- Elisabeth C W van Straaten
- Department of Clinical Neurophysiology, MEG Center, VU Medical Center, Amsterdam, Netherlands; Nutricia Advanced Medical Nutrition, Nutricia Research, Utrecht, Netherlands
| | - Hanneke de Waal
- Department of Neurology, Alzheimer Center, VU Medical Center , Amsterdam , Netherlands
| | | | - Philip Scheltens
- Department of Neurology, Alzheimer Center, VU Medical Center , Amsterdam , Netherlands
| | - Fernando Maestu
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology , Madrid , Spain
| | - Rafal Nowak
- Magnetoencephalography Unit, Centro Medico Teknon , Barcelona , Spain
| | - Arjan Hillebrand
- Department of Clinical Neurophysiology, MEG Center, VU Medical Center , Amsterdam , Netherlands
| | - Cornelis J Stam
- Department of Clinical Neurophysiology, MEG Center, VU Medical Center , Amsterdam , Netherlands
| |
Collapse
|
34
|
Pérez-Cañamás A, Sarroca S, Melero-Jerez C, Porquet D, Sansa J, Knafo S, Esteban JA, Sanfeliu C, Ledesma MD. A diet enriched with plant sterols prevents the memory impairment induced by cholesterol loss in senescence-accelerated mice. Neurobiol Aging 2016; 48:1-12. [PMID: 27622776 DOI: 10.1016/j.neurobiolaging.2016.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 07/07/2016] [Accepted: 08/09/2016] [Indexed: 01/02/2023]
Abstract
Cholesterol reduction at the neuronal plasma membrane has been related to age-dependent cognitive decline. We have used senescent-accelerated mice strain 8 (SAMP8), an animal model for aging, to examine the association between cholesterol loss and cognitive impairment and to test strategies to revert this process. We show that the hippocampus of SAMP8 mice presents reduced cholesterol levels and enhanced amount of its degrading enzyme Cyp46A1 (Cyp46) already at 6 months of age. Cholesterol loss accounts for the impaired long-term potentiation in these mice. Plant sterol (PSE)-enriched diet prevents long-term potentiation impairment and cognitive deficits in SAMP8 mice without altering cholesterol levels. PSE diet also reduces the abnormally high amyloid peptide levels in SAMP8 mice brains and restores membrane compartmentalization of presenilin1, the catalytic component of the amyloidogenic γ-secretase. These results highlight the influence of cholesterol loss in age-related cognitive decline and provide with a noninvasive strategy to counteract it. Our results suggest that PSE overtake cholesterol functions in the brain contributing to reduce deleterious consequences of cholesterol loss during aging.
Collapse
Affiliation(s)
| | - Sara Sarroca
- Institut d'Investigacions Biomèdiques de Barcelona, CSIC, Barcelona, Spain
| | | | - David Porquet
- Institut d'Investigacions Biomèdiques de Barcelona, CSIC, Barcelona, Spain
| | - Joan Sansa
- Departament de Psicologia Bàsica, Universitat de Barcelona, Barcelona, Spain
| | - Shira Knafo
- Centro Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain; Unidad de Biofísica CSIC-UPV/EHU, Campus Universidad del País Vasco, Leioa, Spain; IkerBasque, Basque Foundation for Science, Basque Country, Spain
| | - Jose A Esteban
- Centro Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
| | - Coral Sanfeliu
- Institut d'Investigacions Biomèdiques de Barcelona, CSIC, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain.
| | | |
Collapse
|
35
|
Wu K, Gao X, Shi B, Chen S, Zhou X, Li Z, Gan Y, Cui L, Kang JX, Li W, Huang R. Enriched endogenous n-3 polyunsaturated fatty acids alleviate cognitive and behavioral deficits in a mice model of Alzheimer's disease. Neuroscience 2016; 333:345-55. [PMID: 27474225 DOI: 10.1016/j.neuroscience.2016.07.038] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 07/21/2016] [Accepted: 07/21/2016] [Indexed: 11/29/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that accompanied by memory deficits and neuropsychiatric dysfunction. Omega-3 polyunsaturated fatty acids (n-3 PUFAs) have seemly therapeutic potential in AD, but the benefit of n-3 PUFAs is still in debates. Here, we employed a transgenic mice carry fat-1 gene to encode n-3 desaturase from Caenorhabditis elegans, which increase endogenous n-3 PUFAs by converting n-6 PUFAs to n-3 PUFAs crossed with amyloid precursor protein (APP) Tg mice to evaluate the protective effects of endogenous n-3 PUFAs on cognitive and behavioral deficits of APP Tg mice. We fed APP, APP/fat-1 and fat-1 mice with n-6 PUFAs rich diet. Brain tissues were collected at 3, 9 and 12 months for fatty acid and gene expression analysis, histology and protein assays. Morris Water Maze Test, open field test and elevated plus maze test were performed to measure the behavior capability. From the results, the expression of fat-1 transgene increased cortical n-3: n-6 PUFAs ratio and n-3 PUFAs concentrations, and sensorimotor dysfunction and cognitive deficits in AD were significantly less severe in APP/fat-1 mice with endogenous n-3 PUFAs than in APP mice controls. The protection against disturbance of spontaneous motor activity and cognitive deficits in AD was strongly correlated with increased n-3: n-6 PUFAs ratio and endogenous n-3 PUFAs, reduced APP generation, inhibited amyloid β peptide aggregation, suppressed nuclear factor-kappa B and astroglia activation, and reduced death of neurons in the cortex of APP/fat-1 mice compared with APP mice controls. In conclusion, our study demonstrates that an available medication with the maintenance of enriched n-3 PUFAs in the brain could slow down cognitive decline and prevent neuropsychological disorder in AD.
Collapse
Affiliation(s)
- Kefeng Wu
- Guangdong Key Laboratory for Research and Development of Natural Drug, Guangdong Medical College, Zhanjiang, Guangdong, China
| | - Xiang Gao
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang, China
| | - Baoyan Shi
- Guangdong Key Laboratory for Research and Development of Natural Drug, Guangdong Medical College, Zhanjiang, Guangdong, China; Guangdong Key laboratory of Laboratory Animal, Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China
| | - Shiyu Chen
- Guangdong Key Laboratory for Research and Development of Natural Drug, Guangdong Medical College, Zhanjiang, Guangdong, China; Guangdong Key laboratory of Laboratory Animal, Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China
| | - Xin Zhou
- Guangdong Key Laboratory for Research and Development of Natural Drug, Guangdong Medical College, Zhanjiang, Guangdong, China
| | - Zhidong Li
- Department of Pharmacology, Guangdong Medical College, Zhanjiang, Guangdong 524023, China
| | - Yuhong Gan
- Department of Pharmacology, Guangdong Medical College, Zhanjiang, Guangdong 524023, China
| | - Liao Cui
- Guangdong Key Laboratory for Research and Development of Natural Drug, Guangdong Medical College, Zhanjiang, Guangdong, China; Department of Pharmacology, Guangdong Medical College, Zhanjiang, Guangdong 524023, China
| | - Jing Xuan Kang
- The Laboratory for Lipid Medicine and Technology, Massachusetts General Hospital, Boston 02114, USA
| | - Wende Li
- Guangdong Key Laboratory for Research and Development of Natural Drug, Guangdong Medical College, Zhanjiang, Guangdong, China; Guangdong Key laboratory of Laboratory Animal, Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China.
| | - Ren Huang
- Guangdong Key laboratory of Laboratory Animal, Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China.
| |
Collapse
|
36
|
Hydrogen sulfide down-regulates BACE1 and PS1 via activating PI3K/Akt pathway in the brain of APP/PS1 transgenic mouse. Pharmacol Rep 2016; 68:975-82. [PMID: 27372924 DOI: 10.1016/j.pharep.2016.05.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/12/2016] [Accepted: 05/13/2016] [Indexed: 11/22/2022]
Abstract
BACKGROUND Endogenous hydrogen sulfide (H2S) may have multiple physiological functions in brain. Our previous study showed that H2S improved spatial memory impairment and decreased the production of Aβ in APP/PS1 transgenic mice. However, many of the underlying mechanisms are not still being elucidated. The aim of the present study is to investigate the neuroprotective mechanisms of H2S involving in the activity of β-secretase (BACE1), γ-secretase (PS1) and α-secretase (ADAM17). METHODS Morris water maze was used to measure the behavior change. The levels of Aβ40 and Aβ42 were quantified using colorimetric ELISA kits and immunohistochemical analysis. The levels of BACE1, PS1, ADAM17, pAkt, pp38MAPK, pERK and pJNK were tested by Western blot analysis in normal mice, APP/PS1 transgenic mice and 50μmol/kg-NaHS-treated transgenic mice. On the basis of exogenous H2S treatment, LY294002 (inhibitors of PI3K/Akt) or PD98059 (inhibitors of MAPK/ERK) was injected into lateral cerebral ventricle. RESULTS The levels of BACE1, PS1 and pp38MAPK were increased and ADAM17 were decreased in the APP/PS1 transgenic mice. After intraperitoneal administration of an H2S donor (NaHS) into APP/PS1 mice, the levels of BACE1, PS1 and pp38MAPK were reduced and ADAM17 increased. The level of pp38 MAPKs, pAkt and pERK1/2 was increased in APP/PS1 transgenic mice compared with normal mice (p<0.05). There was no difference in the expression of pJNK between AD transgenic mice and normal mice (p>0.05). These results demonstrated that LY294002 inhibited the effect of H2S on decreasing the BACE1 and PS1, reducing the level of Aβ and improving memory impairment in APP/PS1 transgenic mice. PD98059 had no influence on the expression of BACE1 and PS1. CONCLUSIONS H2S inhibits the expression of BACE1 and PS1 by activating PI3K/Akt pathway in AD.
Collapse
|
37
|
Cansev M. Synaptogenesis: Modulation by Availability of Membrane Phospholipid Precursors. Neuromolecular Med 2016; 18:426-40. [DOI: 10.1007/s12017-016-8414-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 05/24/2016] [Indexed: 10/21/2022]
|
38
|
Belkouch M, Hachem M, Elgot A, Lo Van A, Picq M, Guichardant M, Lagarde M, Bernoud-Hubac N. The pleiotropic effects of omega-3 docosahexaenoic acid on the hallmarks of Alzheimer's disease. J Nutr Biochem 2016; 38:1-11. [PMID: 27825512 DOI: 10.1016/j.jnutbio.2016.03.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/14/2015] [Accepted: 03/03/2016] [Indexed: 12/17/2022]
Abstract
Among omega-3 polyunsaturated fatty acids (PUFAs), docosahexaenoic acid (DHA, 22:6n-3) is important for adequate brain development and cognition. DHA is highly concentrated in the brain and plays an essential role in brain functioning. DHA, one of the major constituents in fish fats, readily crosses the blood-brain barrier from blood to the brain. Its critical role was further supported by its reduced levels in the brain of Alzheimer's disease (AD) patients. This agrees with a potential role of DHA in memory, learning and cognitive processes. Since there is yet no cure for dementia such as AD, there is growing interest in the role of DHA-supplemented diet in the prevention of AD pathogenesis. Accordingly, animal, epidemiological, preclinical and clinical studies indicated that DHA has neuroprotective effects in a number of neurodegenerative conditions including AD. The beneficial effects of this key omega-3 fatty acid supplementation may depend on the stage of disease progression, other dietary mediators and the apolipoprotein ApoE genotype. Herein, our review investigates, from animal and cell culture studies, the molecular mechanisms involved in the neuroprotective potential of DHA with emphasis on AD.
Collapse
Affiliation(s)
- Mounir Belkouch
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France.
| | - Mayssa Hachem
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Abdeljalil Elgot
- Laboratoire des Sciences et Technologies de la Santé, Unité des Sciences Biomédicales, Institut Supérieur des Sciences de la Santé, Université Hassan 1er, Settat, Morocco
| | - Amanda Lo Van
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Madeleine Picq
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Michel Guichardant
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Michel Lagarde
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| | - Nathalie Bernoud-Hubac
- Université de Lyon, UMR INSERM 1060, UMR INRA 1397, IMBL/INSA-Lyon, Cardiovasculaire, Métabolisme, Diabétologie et Nutrition Laboratory, Bât Louis Pasteur, INSA, Villeurbanne, France
| |
Collapse
|
39
|
Koivisto H, Leinonen H, Puurula M, Hafez HS, Barrera GA, Stridh MH, Waagepetersen HS, Tiainen M, Soininen P, Zilberter Y, Tanila H. Chronic Pyruvate Supplementation Increases Exploratory Activity and Brain Energy Reserves in Young and Middle-Aged Mice. Front Aging Neurosci 2016; 8:41. [PMID: 27014054 PMCID: PMC4794631 DOI: 10.3389/fnagi.2016.00041] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 02/15/2016] [Indexed: 01/09/2023] Open
Abstract
Numerous studies have reported neuroprotective effects of pyruvate when given in systemic injections. Impaired glucose uptake and metabolism are found in Alzheimer's disease (AD) and in AD mouse models. We tested whether dietary pyruvate supplementation is able to provide added energy supply to brain and thereby attenuate aging- or AD-related cognitive impairment. Mice received ~800 mg/kg/day Na-pyruvate in their chow for 2-6 months. In middle-aged wild-type mice and in 6.5-month-old APP/PS1 mice, pyruvate facilitated spatial learning and increased exploration of a novel odor. However, in passive avoidance task for fear memory, the treatment group was clearly impaired. Independent of age, long-term pyruvate increased explorative behavior, which likely explains the paradoxical impairment in passive avoidance. We also assessed pyruvate effects on body weight, muscle force, and endurance, and found no effects. Metabolic postmortem assays revealed increased energy compounds in nuclear magnetic resonance spectroscopy as well as increased brain glycogen storages in the pyruvate group. Pyruvate supplementation may counteract aging-related behavioral impairment, but its beneficial effect seems related to increased explorative activity rather than direct memory enhancement.
Collapse
Affiliation(s)
| | - Henri Leinonen
- A. I. Virtanen Institute, University of Eastern Finland , Kuopio , Finland
| | - Mari Puurula
- A. I. Virtanen Institute, University of Eastern Finland , Kuopio , Finland
| | - Hani Sayed Hafez
- Biology Department, Faculty of Science, Suez University , Suez , Egypt
| | | | - Malin H Stridh
- Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Helle S Waagepetersen
- Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Mika Tiainen
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland , Kuopio , Finland
| | - Pasi Soininen
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland , Kuopio , Finland
| | - Yuri Zilberter
- Institut national de la santé et de la recherche médicale UMR_S 1106, Institut de Neurosciences des Systèmes, Aix-Marseille Université , Marseille , France
| | - Heikki Tanila
- A. I. Virtanen Institute, University of Eastern Finland , Kuopio , Finland
| |
Collapse
|
40
|
Ottaviano G, Frasson G, Nardello E, Martini A. Olfaction deterioration in cognitive disorders in the elderly. Aging Clin Exp Res 2016; 28:37-45. [PMID: 26003671 DOI: 10.1007/s40520-015-0380-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 05/12/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Parkinson's and Alzheimer's diseases are widespread neurodegenerative pathologies. Parkinson's disease affects about 1 % of the population over the age of 65 years, while Alzheimer is considered the most common cause of dementia, with an annual incidence of 1 % in persons aged 65 years. It has been demonstrated that both these neurodegenerative diseases are associated with smell dysfunction. AIM The aim of the present review is to describe briefly modern olfactory evaluation tools as well as the importance of olfactory sensitivity screening in the elderly, especially where cognitive disorders, such as Alzheimer's or Parkinson's diseases, are suspected. METHODS A brief literature review focusing on the basic principle of smell tests is illustrated together with their application in elderly patients affected by cognitive disorders, in particular Parkinson's and Alzheimer's diseases. RESULTS/CONCLUSIONS Alzheimer's and Parkinson's diseases are both neurodegenerative disorders typically found in the elderly. As both diseases are characterized by the early presence of dysosmia, simple validated smell tests could very well help clinicians in the early diagnosis of these neuropathological conditions. Elderly patients complaining of smell loss and found to be dysosmic, by means of validated olfactory tests, should be neurologically evaluated as early as possible to detect slight motor abnormalities in an at-risk population.
Collapse
|
41
|
Poisson LM, Suhail H, Singh J, Datta I, Denic A, Labuzek K, Hoda MN, Shankar A, Kumar A, Cerghet M, Elias S, Mohney RP, Rodriguez M, Rattan R, Mangalam AK, Giri S. Untargeted Plasma Metabolomics Identifies Endogenous Metabolite with Drug-like Properties in Chronic Animal Model of Multiple Sclerosis. J Biol Chem 2015; 290:30697-712. [PMID: 26546682 DOI: 10.1074/jbc.m115.679068] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Indexed: 12/20/2022] Open
Abstract
We performed untargeted metabolomics in plasma of B6 mice with experimental autoimmune encephalitis (EAE) at the chronic phase of the disease in search of an altered metabolic pathway(s). Of 324 metabolites measured, 100 metabolites that mapped to various pathways (mainly lipids) linked to mitochondrial function, inflammation, and membrane stability were observed to be significantly altered between EAE and control (p < 0.05, false discovery rate <0.10). Bioinformatics analysis revealed six metabolic pathways being impacted and altered in EAE, including α-linolenic acid and linoleic acid metabolism (PUFA). The metabolites of PUFAs, including ω-3 and ω-6 fatty acids, are commonly decreased in mouse models of multiple sclerosis (MS) and in patients with MS. Daily oral administration of resolvin D1, a downstream metabolite of ω-3, decreased disease progression by suppressing autoreactive T cells and inducing an M2 phenotype of monocytes/macrophages and resident brain microglial cells. This study provides a proof of principle for the application of metabolomics to identify an endogenous metabolite(s) possessing drug-like properties, which is assessed for therapy in preclinical mouse models of MS.
Collapse
Affiliation(s)
- Laila M Poisson
- From the Center for Bioinformatics and Departments of Public Health Sciences and
| | | | | | - Indrani Datta
- From the Center for Bioinformatics and Departments of Public Health Sciences and
| | | | - Krzysztof Labuzek
- the Department of Pharmacology, Medical University of Silesia, Medyków 18, PL 40-752 Katowice, Poland
| | - Md Nasrul Hoda
- the Department of Neurology, Georgia Health Sciences University, Augusta, Georgia 30912, the Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia 30912
| | | | - Ashok Kumar
- the Department of Anatomy and Cell Biology, School of Medicine, Wayne State University, Detroit, Michigan 48202
| | | | | | | | - Moses Rodriguez
- the Departments of Neurology and Immunology, Mayo Clinic College of Medicine, Rochester, Minnesota 55906
| | - Ramandeep Rattan
- Division of Gynecology Oncology, Department of Women's Health Services, Henry Ford Health System, Detroit, Michigan 48202
| | - Ashutosh K Mangalam
- the Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | | |
Collapse
|
42
|
Teng E, Taylor K, Bilousova T, Weiland D, Pham T, Zuo X, Yang F, Chen PP, Glabe CG, Takacs A, Hoffman DR, Frautschy SA, Cole GM. Dietary DHA supplementation in an APP/PS1 transgenic rat model of AD reduces behavioral and Aβ pathology and modulates Aβ oligomerization. Neurobiol Dis 2015; 82:552-560. [PMID: 26369878 DOI: 10.1016/j.nbd.2015.09.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 08/22/2015] [Accepted: 09/07/2015] [Indexed: 12/18/2022] Open
Abstract
Increased dietary consumption of docosahexaenoic acid (DHA) is associated with decreased risk for Alzheimer's disease (AD). These effects have been postulated to arise from DHA's pleiotropic effects on AD pathophysiology, including its effects on β-amyloid (Aβ) production, aggregation, and toxicity. While in vitro studies suggest that DHA may inhibit and reverse the formation of toxic Aβ oligomers, it remains uncertain whether these mechanisms operate in vivo at the physiological concentrations of DHA attainable through dietary supplementation. We sought to clarify the effects of dietary DHA supplementation on Aβ indices in a transgenic APP/PS1 rat model of AD. Animals maintained on a DHA-supplemented diet exhibited reductions in hippocampal Aβ plaque density and modest improvements on behavioral testing relative to those maintained on a DHA-depleted diet. However, DHA supplementation also increased overall soluble Aβ oligomer levels in the hippocampus. Further quantification of specific conformational populations of Aβ oligomers indicated that DHA supplementation increased fibrillar (i.e. putatively less toxic) Aβ oligomers and decreased prefibrillar (i.e. putatively more toxic) Aβ oligomers. These results provide in vivo evidence suggesting that DHA can modulate Aβ aggregation by stabilizing soluble fibrillar Aβ oligomers and thus reduce the formation of both Aβ plaques and prefibrillar Aβ oligomers. However, since fibrillar Aβ oligomers still retain inherent neurotoxicity, DHA may need to be combined with other interventions that can additionally reduce fibrillar Aβ oligomer levels for more effective prevention of AD in clinical settings.
Collapse
Affiliation(s)
- Edmond Teng
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA.
| | - Karen Taylor
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Tina Bilousova
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - David Weiland
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Thaidan Pham
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Xiaohong Zuo
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA; Department of Neurobiology and Neurology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Fusheng Yang
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Ping-Ping Chen
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Charles G Glabe
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA; Biochemistry Department and Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Dennis R Hoffman
- Retina Foundation of the Southwest, Dallas, TX, USA; Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sally A Frautschy
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Gregory M Cole
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
43
|
Vanmierlo T, Bogie JF, Mailleux J, Vanmol J, Lütjohann D, Mulder M, Hendriks JJ. Plant sterols: Friend or foe in CNS disorders? Prog Lipid Res 2015; 58:26-39. [DOI: 10.1016/j.plipres.2015.01.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 01/15/2015] [Accepted: 01/15/2015] [Indexed: 12/21/2022]
|
44
|
A specific multi-nutrient enriched diet enhances hippocampal cholinergic transmission in aged rats. Neurobiol Aging 2015; 36:344-51. [DOI: 10.1016/j.neurobiolaging.2014.07.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 06/23/2014] [Accepted: 07/18/2014] [Indexed: 02/02/2023]
|
45
|
Smiljanic K, Vanmierlo T, Mladenovic Djordjevic A, Perovic M, Ivkovic S, Lütjohann D, Kanazir S. Cholesterol metabolism changes under long-term dietary restrictions while the cholesterol homeostasis remains unaffected in the cortex and hippocampus of aging rats. AGE (DORDRECHT, NETHERLANDS) 2014; 36:9654. [PMID: 24756765 PMCID: PMC4082575 DOI: 10.1007/s11357-014-9654-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 04/05/2014] [Indexed: 06/03/2023]
Abstract
Maintaining cholesterol homeostasis in the brain is vital for its proper functioning. While it is well documented that dietary restriction modulates the metabolism of cholesterol peripherally, little is known as to how it can affect cholesterol metabolism in the brain. The present study was designed to elucidate the impact of long-term dietary restriction on brain cholesterol metabolism. Three-month-old male Wistar rats were exposed to long-term dietary restriction until 12 and 24 months of age. The concentrations of cholesterol, its precursors and metabolites, and food-derived phytosterols were measured in the serum, cortex, and hippocampus by gas chromatography/mass spectrometry. Relative changes in the levels of proteins involved in cholesterol synthesis, transport, and degradation were determined by Western blot analysis. Reduced food intake influenced the expression patterns of proteins implicated in cholesterol metabolism in the brain in a region-specific manner. Dietary restriction decreased the concentrations of cholesterol precursors, lanosterol in the cortex, and lanosterol and lathosterol in the hippocampus at 12 months, while the level of desmosterol was elevated in the hippocampus at 24 months. The concentrations of cholesterol and 24(S)-hydroxycholesterol remained unaffected. Food-derived phytosterols were significantly lower after dietary restriction in both the cortex and hippocampus at 12 and 24 months. These findings provide new insight into the effects of dietary restriction on cholesterol metabolism in the brain, lending further support to its neuroprotective effect.
Collapse
Affiliation(s)
- Kosara Smiljanic
- />Department of Neurobiology, Institute for Biological Research, University of Belgrade, Bul. despota Stefana 142, 11060 Belgrade, Serbia
| | - Tim Vanmierlo
- />Department of Immunology and Biochemistry, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Aleksandra Mladenovic Djordjevic
- />Department of Neurobiology, Institute for Biological Research, University of Belgrade, Bul. despota Stefana 142, 11060 Belgrade, Serbia
| | - Milka Perovic
- />Department of Neurobiology, Institute for Biological Research, University of Belgrade, Bul. despota Stefana 142, 11060 Belgrade, Serbia
| | - Sanja Ivkovic
- />Department of Developmental Biology, Institute for Molecular Medicine, Lisbon, Portugal
| | - Dieter Lütjohann
- />Laboratory for Special Lipid Diagnostics, Institute of Clinical Chemistry and Clinical Pharmacology, University Clinics of Bonn, Bonn, Germany
| | - Selma Kanazir
- />Department of Neurobiology, Institute for Biological Research, University of Belgrade, Bul. despota Stefana 142, 11060 Belgrade, Serbia
| |
Collapse
|