1
|
Huang H, Apaijai N, Oo TT, Suntornsaratoon P, Charoenphandhu N, Chattipakorn N, Chattipakorn SC. Gestational diabetes mellitus, not obesity, triggers postpartum brain inflammation and premature aging in Sprague-Dawley rats. Neuroscience 2024; 559:166-180. [PMID: 39236804 DOI: 10.1016/j.neuroscience.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/10/2024] [Accepted: 09/01/2024] [Indexed: 09/07/2024]
Abstract
Previous studies showed that women with gestational diabetes mellitus (GDM) are susceptible to cognitive dysfunction. We investigated the effects of GDM on brain pathologies and premature brain aging in rats. Seven-week-old female Sprague-Dawley rats were fed a normal diet (ND) or a high-fat diet (HFD) after two weeks of acclimatization. On pregnancy day 0, HFD-treated rats received streptozotocin (GDM group) or vehicle (Obese mothers). ND-treated rats received vehicle (ND-control mothers). On postpartum day 21, brains and blood were collected. The GDM group showed increased inflammatory and premature aging markers, mitochondrial changes, and compensatory increases in the blood-brain barrier and synaptic proteins in the prefrontal cortex and hippocampus. GDM triggers maternal brain inflammation and premature aging, suggesting compensatory mechanisms may protect against these effects.
Collapse
Affiliation(s)
- Huatuo Huang
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand; Center for Medical Laboratory Science, the Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Nattayaporn Apaijai
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand
| | - Thura Tun Oo
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand
| | - Panan Suntornsaratoon
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand; Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Narattaphol Charoenphandhu
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand; Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand; Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand; The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
2
|
Yue Q, Leng X, Xie N, Zhang Z, Yang D, Hoi MPM. Endothelial Dysfunctions in Blood-Brain Barrier Breakdown in Alzheimer's Disease: From Mechanisms to Potential Therapies. CNS Neurosci Ther 2024; 30:e70079. [PMID: 39548663 PMCID: PMC11567945 DOI: 10.1111/cns.70079] [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: 07/17/2024] [Revised: 09/13/2024] [Accepted: 09/28/2024] [Indexed: 11/18/2024] Open
Abstract
Recent research has shown the presence of blood-brain barrier (BBB) breakdown in Alzheimer's disease (AD). BBB is a dynamic interface consisting of a continuous monolayer of brain endothelial cells (BECs) enveloped by pericytes and astrocytes. The restricted permeability of BBB strictly controls the exchange of substances between blood and brain parenchyma, which is crucial for brain homeostasis by excluding blood-derived detrimental factors and pumping out brain-derived toxic molecules. BBB breakdown in AD is featured as a series of BEC pathologies such as increased paracellular permeability, abnormal levels and functions of transporters, and inflammatory or oxidative profile, which may disturb the substance transportation across BBB, thereafter induce CNS disorders such as hypometabolism, Aβ accumulation, and neuroinflammation, eventually aggravate cognitive decline. Therefore, it seems important to protect BEC properties for BBB maintenance and neuroprotection. In this review, we thoroughly summarized the pathological alterations of BEC properties reported in AD patients and numerous AD models, including paracellular permeability, influx and efflux transporters, and inflammatory and oxidative profiles, and probably associated underlying mechanisms. Then we reviewed current therapeutic agents that are effective in ameliorating a series of BEC pathologies, and ultimately protecting BBB integrity and cognitive functions. Regarding the current drug development for AD proceeds extremely hard, this review aims to discuss the therapeutic potentials of targeting BEC pathologies and BBB maintenance for AD treatment, therefore expecting to shed a light on the future AD drug development by targeting BEC pathologies and BBB protection.
Collapse
Affiliation(s)
- Qian Yue
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacao SARChina
- Department of Pharmaceutical Sciences, Faculty of Health SciencesUniversity of MacauMacao SARChina
- Department of CardiologyThe First Affiliated Hospital of Jinan UniversityGuangzhouGuangdongChina
- The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital)HeyuanGuangdongChina
| | - Xinyue Leng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacao SARChina
- Department of Pharmaceutical Sciences, Faculty of Health SciencesUniversity of MacauMacao SARChina
| | - Ningqing Xie
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio‐Cerebrovascular Diseases, and Institute of New Drug ResearchJinan UniversityGuangzhouChina
- Guangdong‐Hong Kong‐Macau Joint Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs ResearchJinan University College of PharmacyGuangzhouChina
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)Jinan University College of PharmacyGuangzhouChina
| | - Zaijun Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio‐Cerebrovascular Diseases, and Institute of New Drug ResearchJinan UniversityGuangzhouChina
- Guangdong‐Hong Kong‐Macau Joint Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs ResearchJinan University College of PharmacyGuangzhouChina
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)Jinan University College of PharmacyGuangzhouChina
| | - Deguang Yang
- Department of CardiologyThe First Affiliated Hospital of Jinan UniversityGuangzhouGuangdongChina
- The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital)HeyuanGuangdongChina
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)Jinan University College of PharmacyGuangzhouChina
| | - Maggie Pui Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacao SARChina
- Department of Pharmaceutical Sciences, Faculty of Health SciencesUniversity of MacauMacao SARChina
| |
Collapse
|
3
|
Almanza DLV, Koletar MM, Lai AY, Lam WW, Joo L, Hill ME, Stanisz GJ, McLaurin J, Stefanovic B. High caloric intake improves neuronal metabolism and functional hyperemia in a rat model of early AD pathology. Theranostics 2024; 14:7405-7423. [PMID: 39659583 PMCID: PMC11626934 DOI: 10.7150/thno.98793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 10/24/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction: While obesity has been linked to both increased and decreased rate of cognitive decline in Alzheimer's Disease (AD) patients, there is no consensus on the interaction between obesity and AD. Methods: The TgF344-AD rat model was used to investigate the effects of high carbohydrate, high fat (HCHF) diet on brain glucose metabolism and hemodynamics in the presence or absence of AD transgenes, in presymptomatic (6-month-old) vs. symptomatic (12-month-old) stages of AD progression using non-invasive neuroimaging. Results: In presymptomatic AD, HCHF exerted detrimental effects, attenuating both hippocampal glucose uptake and resting perfusion in both non-transgenic and TgAD cohorts, when compared to CHOW-fed cohorts. In contrast, HCHF consumption was beneficial in established AD, resolving the AD-progression associated attenuation in hippocampal glucose uptake and functional hyperemia. Discussion: Whereas HCHF was harmful to the presymptomatic AD brain, it ameliorated deficits in hippocampal metabolism and neurovascular coupling in symptomatic TgAD rats.
Collapse
Affiliation(s)
- Dustin Loren V. Almanza
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | | | - Aaron Y. Lai
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Wilfred W. Lam
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Lewis Joo
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Mary E. Hill
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Greg J. Stanisz
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University, Lublin, Poland
| | - JoAnne McLaurin
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Bojana Stefanovic
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|
4
|
Wen W, Huang SM, Zhang B. Mechanisms Underlying Obesity-induced Aβ Accumulation in Alzheimer's Disease: A Qualitative Review. J Integr Neurosci 2024; 23:163. [PMID: 39344225 DOI: 10.31083/j.jin2309163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/27/2024] [Accepted: 03/05/2024] [Indexed: 10/01/2024] Open
Abstract
Epidemiological studies show that individuals with obesity are more likely to develop Alzheimer's disease (AD) than those who do not have obesity. However, the mechanisms underlying the relationship between obesity and AD are not entirely unclear. Here, we have reviewed and analyzed relevant articles published in the literature and found that obesity has correlation or potential increase in the levels of β-amyloid (Aβ) protein, which may explain why people with obesity are more likely to suffer from AD. Additionally, the published findings point to the roles of obesity-related metabolic disorders, such as diabetes, inflammation, oxidative stress, and imbalance in gut microbiota in Aβ accumulation caused by obesity. Therefore, in-depth experimental and clinical studies on these mechanisms in the future may help shed light on appropriate prevention and treatment strategies for AD, such as dietary changes and regular exercise to reverse or prevent obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Wei Wen
- Department of Pharmacology, College of Basic Medicine, Heilongjiang University of Chinese Medicine, 150040 Harbin, Heilongjiang, China
| | - Shu-Ming Huang
- Department of Neuroscience, Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, 150040 Harbin, Heilongjiang, China
| | - Bo Zhang
- Department of Neuroscience, Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, 150040 Harbin, Heilongjiang, China
| |
Collapse
|
5
|
Han J, Wang Y, Wei P, Lu D, Shan Y. Unveiling the hidden connection: the blood-brain barrier's role in epilepsy. Front Neurol 2024; 15:1413023. [PMID: 39206290 PMCID: PMC11349696 DOI: 10.3389/fneur.2024.1413023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/18/2024] [Indexed: 09/04/2024] Open
Abstract
Epilepsy is characterized by abnormal synchronous electrical activity of neurons in the brain. The blood-brain barrier, which is mainly composed of endothelial cells, pericytes, astrocytes and other cell types and is formed by connections between a variety of cells, is the key physiological structure connecting the blood and brain tissue and is critical for maintaining the microenvironment in the brain. Physiologically, the blood-brain barrier controls the microenvironment in the brain mainly by regulating the passage of various substances. Disruption of the blood-brain barrier and increased leakage of specific substances, which ultimately leading to weakened cell junctions and abnormal regulation of ion concentrations, have been observed during the development and progression of epilepsy in both clinical studies and animal models. In addition, disruption of the blood-brain barrier increases drug resistance through interference with drug trafficking mechanisms. The changes in the blood-brain barrier in epilepsy mainly affect molecular pathways associated with angiogenesis, inflammation, and oxidative stress. Further research on biomarkers is a promising direction for the development of new therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | - Yongzhi Shan
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Rhea EM, Leclerc M, Yassine HN, Capuano AW, Tong H, Petyuk VA, Macauley SL, Fioramonti X, Carmichael O, Calon F, Arvanitakis Z. State of the Science on Brain Insulin Resistance and Cognitive Decline Due to Alzheimer's Disease. Aging Dis 2024; 15:1688-1725. [PMID: 37611907 PMCID: PMC11272209 DOI: 10.14336/ad.2023.0814] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/14/2023] [Indexed: 08/25/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is common and increasing in prevalence worldwide, with devastating public health consequences. While peripheral insulin resistance is a key feature of most forms of T2DM and has been investigated for over a century, research on brain insulin resistance (BIR) has more recently been developed, including in the context of T2DM and non-diabetes states. Recent data support the presence of BIR in the aging brain, even in non-diabetes states, and found that BIR may be a feature in Alzheimer's disease (AD) and contributes to cognitive impairment. Further, therapies used to treat T2DM are now being investigated in the context of AD treatment and prevention, including insulin. In this review, we offer a definition of BIR, and present evidence for BIR in AD; we discuss the expression, function, and activation of the insulin receptor (INSR) in the brain; how BIR could develop; tools to study BIR; how BIR correlates with current AD hallmarks; and regional/cellular involvement of BIR. We close with a discussion on resilience to both BIR and AD, how current tools can be improved to better understand BIR, and future avenues for research. Overall, this review and position paper highlights BIR as a plausible therapeutic target for the prevention of cognitive decline and dementia due to AD.
Collapse
Affiliation(s)
- Elizabeth M Rhea
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA.
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA 98195, USA.
| | - Manon Leclerc
- Faculty of Pharmacy, Laval University, Quebec, Quebec, Canada.
- Neuroscience Axis, CHU de Québec Research Center - Laval University, Quebec, Quebec, Canada.
| | - Hussein N Yassine
- Departments of Neurology and Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Ana W Capuano
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Han Tong
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Vladislav A Petyuk
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA.
| | - Shannon L Macauley
- Department of Physiology, University of Kentucky, Lexington, KY 40508, USA.
| | - Xavier Fioramonti
- International Associated Laboratory OptiNutriBrain, Bordeaux, France and Quebec, Canada.
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France.
| | - Owen Carmichael
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| | - Frederic Calon
- Faculty of Pharmacy, Laval University, Quebec, Quebec, Canada.
- Neuroscience Axis, CHU de Québec Research Center - Laval University, Quebec, Quebec, Canada.
- International Associated Laboratory OptiNutriBrain, Bordeaux, France and Quebec, Canada.
| | - Zoe Arvanitakis
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
7
|
Ahmed M, Lai AY, Hill ME, Ribeiro JA, Amiraslani A, McLaurin J. Obesity differentially effects the somatosensory cortex and striatum of TgF344-AD rats. Sci Rep 2024; 14:7235. [PMID: 38538727 PMCID: PMC10973391 DOI: 10.1038/s41598-024-57953-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024] Open
Abstract
Lifestyle choices leading to obesity, hypertension and diabetes in mid-life contribute directly to the risk of late-life Alzheimer's disease (AD). However, in late-life or in late-stage AD conditions, obesity reduces the risk of AD and disease progression. To examine the mechanisms underlying this paradox, TgF344-AD rats were fed a varied high-carbohydrate, high-fat (HCHF) diet to induce obesity from nine months of age representing early stages of AD to twelve months of age in which rats exhibit the full spectrum of AD symptomology. We hypothesized regions primarily composed of gray matter, such as the somatosensory cortex (SSC), would be differentially affected compared to regions primarily composed of white matter, such as the striatum. We found increased myelin and oligodendrocytes in the somatosensory cortex of rats fed the HCHF diet with an absence of neuronal loss. We observed decreased inflammation in the somatosensory cortex despite increased AD pathology. Compared to the somatosensory cortex, the striatum had fewer changes. Overall, our results suggest that the interaction between diet and AD progression affects myelination in a brain region specific manner such that regions with a lower density of white matter are preferentially affected. Our results offer a possible mechanistic explanation for the obesity paradox.
Collapse
Affiliation(s)
- Minhal Ahmed
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Aaron Y Lai
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | - Mary E Hill
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | - Jessica A Ribeiro
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | - Ashley Amiraslani
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - JoAnne McLaurin
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada.
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
8
|
Kang L, Wang W, Nie Z, Gong Q, Yao L, Xiang D, Zhang N, Tu N, Feng H, Zong X, Bai H, Wang G, Wang F, Bu L, Liu Z. Dysregulated cerebral blood flow, rather than gray matter Volume, exhibits stronger correlations with blood inflammatory and lipid markers in depression. Neuroimage Clin 2024; 41:103581. [PMID: 38430800 PMCID: PMC10944186 DOI: 10.1016/j.nicl.2024.103581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/08/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024]
Abstract
Arterial spin labeling (ASL) can be used to detect differences in perfusion for multiple brain regions thought to be important in major depressive disorder (MDD). However, the potential of cerebral blood flow (CBF) to predict MDD and its correlations between the blood lipid levels and immune markers, which are closely related to MDD and brain function change, remain unclear. The 451 individuals - 298 with MDD and 133 healthy controls who underwent MRI at a single time point with arterial spin labelling and a high resolution T1-weighted structural scan. A proportion of MDD also provided blood samples for analysis of lipid and immune markers. We performed CBF case-control comparisons, random forest model construction, and exploratory correlation analyses. Moreover, we investigated the relationship between gray matter volume (GMV), blood lipids, and the immune system within the same sample to assess the differences in CBF and GMV. We found that the left inferior parietal but supramarginal and angular gyrus were significantly different between the MDD patients and HCs (voxel-wise P < 0.001, cluster-wise FWE correction). And bilateral inferior temporal (ITG), right middle temporal gyrus and left precentral gyrus CBF predict MDD (the area under the receiver operating characteristic curve of the random forest model is 0.717) and that CBF is a more sensitive predictor of MDD than GMV. The left ITG showed a positive correlation trend with immunoglobulin G (r = 0.260) and CD4 counts (r = 0.283). The right ITG showed a correlation trend with Total Cholesterol (r = -0.249) and tumour necrosis factor-alpha (r = -0.295). Immunity and lipids were closely related to CBF change, with the immunity relationship potentially playing a greater role. The interactions between CBF, plasma lipids and immune index could therefore represent an MDD pathophysiological mechanism. The current findings provide evidence for targeted regulation of CBF or immune properties in MDD.
Collapse
Affiliation(s)
- Lijun Kang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhaowen Nie
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qian Gong
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lihua Yao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dan Xiang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Nan Zhang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ning Tu
- PET/CT/MRI and Molecular Imaging Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongyan Feng
- PET/CT/MRI and Molecular Imaging Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaofen Zong
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hanping Bai
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fei Wang
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China; Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Lihong Bu
- PET/CT/MRI and Molecular Imaging Center, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
9
|
Ahmed M, Lai AY, Hill ME, Ribeiro JA, Amiraslani A, McLaurin J. Obesity differentially effects the somatosensory cortex and striatum of TgF344-AD rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.576454. [PMID: 38545621 PMCID: PMC10970715 DOI: 10.1101/2024.01.22.576454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Lifestyle choices leading to obesity, hypertension and diabetes in mid-life contribute directly to the risk of late-life Alzheimer's disease (AD). However, in late-life or in late-stage AD conditions, obesity reduces the risk of AD and disease progression. To examine the mechanisms underlying this paradox, TgF344-AD rats were fed a varied high-carbohydrate, high-fat (HCHF) diet to induce obesity from nine months of age representing early stages of AD to twelve months of age in which rats exhibit the full spectrum of AD symptomology. We hypothesized regions primarily composed of gray matter, such as the somatosensory cortex (SSC), would be differentially affected compared to regions primarily composed of white matter, such as the striatum. We found increased myelin and oligodendrocytes in the somatosensory cortex of rats fed the HCHF diet with an absence of neuronal loss. We observed decreased inflammation in the somatosensory cortex despite increased AD pathology. Compared to the somatosensory cortex, the striatum had fewer changes. Overall, our results suggest that the interaction between diet and AD progression affects myelination in a brain region specific manner such that regions with a lower density of white matter are preferentially effected. Our results offer a possible mechanistic explanation for the obesity paradox.
Collapse
|
10
|
Li X, Quan M, Wei Y, Wang W, Xu L, Wang Q, Jia J. Critical thinking of Alzheimer's transgenic mouse model: current research and future perspective. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2711-2754. [PMID: 37480469 DOI: 10.1007/s11427-022-2357-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/23/2023] [Indexed: 07/24/2023]
Abstract
Transgenic models are useful tools for studying the pathogenesis of and drug development for Alzheimer's Disease (AD). AD models are constructed usually using overexpression or knock-in of multiple pathogenic gene mutations from familial AD. Each transgenic model has its unique behavioral and pathological features. This review summarizes the research progress of transgenic mouse models, and their progress in the unique mechanism of amyloid-β oligomers, including the first transgenic mouse model built in China based on a single gene mutation (PSEN1 V97L) found in Chinese familial AD. We further summarized the preclinical findings of drugs using the models, and their future application in exploring the upstream mechanisms and multitarget drug development in AD.
Collapse
Affiliation(s)
- Xinyue Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Meina Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Yiping Wei
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Wei Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Lingzhi Xu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Qi Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China.
- Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, 100053, China.
- Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, 100053, China.
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100053, China.
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China.
| |
Collapse
|
11
|
Yehuda H, Madrer N, Goldberg D, Soreq H, Meerson A. Inversely Regulated Inflammation-Related Processes Mediate Anxiety-Obesity Links in Zebrafish Larvae and Adults. Cells 2023; 12:1794. [PMID: 37443828 PMCID: PMC10341043 DOI: 10.3390/cells12131794] [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: 06/14/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Anxiety and metabolic impairments are often inter-related, but the underlying mechanisms are unknown. To seek RNAs involved in the anxiety disorder-metabolic disorder link, we subjected zebrafish larvae to caffeine-induced anxiety or high-fat diet (HFD)-induced obesity followed by RNA sequencing and analyses. Notably, differentially expressed (DE) transcripts in these larval models and an adult zebrafish caffeine-induced anxiety model, as well as the transcript profiles of inherently anxious versus less anxious zebrafish strains and high-fat diet-fed versus standard diet-fed adult zebrafish, revealed inversely regulated DE transcripts. In both larval anxiety and obesity models, these included long noncoding RNAs and transfer RNA fragments, with the overrepresented immune system and inflammation pathways, e.g., the "interleukin signaling pathway" and "inflammation mediated by chemokine and cytokine signaling pathway". In adulthood, overrepresented immune system processes included "T cell activation", "leukocyte cell-cell adhesion", and "antigen processing and presentation". Furthermore, unlike adult zebrafish, obesity in larvae was not accompanied by anxiety-like behavior. Together, these results may reflect an antagonistic pleiotropic phenomenon involving a re-adjusted modulation of the anxiety-metabolic links with an occurrence of the acquired immune system. Furthermore, the HFD potential to normalize anxiety-upregulated immune-related genes may reflect the high-fat diet protection of anxiety and neurodegeneration reported by others.
Collapse
Affiliation(s)
- Hila Yehuda
- MIGAL—Galilee Research Institute, Kiryat Shmona 11016, Israel
- The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; (H.Y.); (N.M.)
| | - Nimrod Madrer
- The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; (H.Y.); (N.M.)
- The Edmond and Lily Safra Center for Brain Science, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Doron Goldberg
- MIGAL—Galilee Research Institute, Kiryat Shmona 11016, Israel
- Tel-Hai College, Upper Galilee 1220800, Israel;
| | - Hermona Soreq
- The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; (H.Y.); (N.M.)
- The Edmond and Lily Safra Center for Brain Science, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Ari Meerson
- MIGAL—Galilee Research Institute, Kiryat Shmona 11016, Israel
- Tel-Hai College, Upper Galilee 1220800, Israel;
| |
Collapse
|
12
|
The Role of Dietary Lipids in Cognitive Health: Implications for Neurodegenerative Disease. Biomedicines 2022; 10:biomedicines10123250. [PMID: 36552006 PMCID: PMC9775642 DOI: 10.3390/biomedicines10123250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022] Open
Abstract
Neurodegenerative diseases are a group of disorders characterised by progressive loss of brain function. The most common of these is Alzheimer's disease, a form of dementia. Intake of macro- and micro-nutrients impacts brain function, including memory, learning, mood, and behaviour. Lipids, particularly phospholipids and sphingolipids, are crucial structural components of neural tissues and significantly affect cognitive function. The importance of functional foods in preventing cardiovascular disease is well-documented in the current literature. However, the significance of such foods for central nervous system health and neurodegenerative diseases is less recognized. Gut microbiome composition affects cognitive health and function, and dietary lipids are known to influence gut health. Thus, this review will discuss different sources of dietary lipids and their effect on cognitive functioning and their interaction with the gut microbiome in the context of neurodegenerative disease.
Collapse
|
13
|
Leikin-Frenkel A, Schnaider Beeri M, Cooper I. How Alpha Linolenic Acid May Sustain Blood-Brain Barrier Integrity and Boost Brain Resilience against Alzheimer's Disease. Nutrients 2022; 14:nu14235091. [PMID: 36501121 PMCID: PMC9737216 DOI: 10.3390/nu14235091] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/08/2022] [Accepted: 11/29/2022] [Indexed: 12/04/2022] Open
Abstract
Cognitive decline, the primary clinical phenotype of Alzheimer's disease (AD), is currently attributed mainly to amyloid and tau protein deposits. However, a growing body of evidence is converging on brain lipids, and blood-brain barrier (BBB) dysfunction, as crucial players involved in AD development. The critical role of lipids metabolism in the brain and its vascular barrier, and its constant modifications particularly throughout AD development, warrants investigation of brain lipid metabolism as a high value therapeutic target. Yet, there is limited knowledge on the biochemical and structural roles of lipids in BBB functionality in AD. Within this framework, we hypothesize that the ApoE4 genotype, strongly linked to AD risk and progression, may be related to altered fatty acids composition in the BBB. Interestingly, alpha linolenic acid (ALA), the precursor of the majoritarian brain component docosahexaenoic acid (DHA), emerges as a potential novel brain savior, acting via BBB functional improvements, and this may be primarily relevant to ApoE4 carriers.
Collapse
Affiliation(s)
- Alicia Leikin-Frenkel
- Bert Strassburger Lipid Center, Sheba Medical Center, Tel-Hashomer 52621, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Michal Schnaider Beeri
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Ramat-Gan 52621, Israel
- Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Itzik Cooper
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Ramat-Gan 52621, Israel
- School of Psychology, The Reichman University (IDC), Herzliya 4610101, Israel
- Correspondence: ; Tel.: +972-3-5303693
| |
Collapse
|
14
|
Stefaniak O, Dobrzyńska M, Drzymała-Czyż S, Przysławski J. Diet in the Prevention of Alzheimer's Disease: Current Knowledge and Future Research Requirements. Nutrients 2022; 14:4564. [PMID: 36364826 PMCID: PMC9656789 DOI: 10.3390/nu14214564] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/23/2022] [Accepted: 10/25/2022] [Indexed: 08/13/2023] Open
Abstract
Alzheimer's disease is a progressive brain disease that is becoming a major health problem in today's world due to the aging population. Despite it being widely known that diet has a significant impact on the prevention and progression of Alzheimer's disease, the literature data are still scarce and controversial. The application of the principles of rational nutrition for the elderly is suggested for Alzheimer's disease. The diet should be rich in neuroprotective nutrients, i.e., antioxidants, B vitamins, and polyunsaturated fatty acids. Some studies suggest that diets such as the Mediterranean diet, the DASH (Dietary Approaches to Stop Hypertension) diet, and the MIND (Mediterranean-DASH Intervention for Neurodegenerative Delay) diet have a beneficial effect on the risk of developing Alzheimer's disease.
Collapse
Affiliation(s)
| | - Małgorzata Dobrzyńska
- Department of Bromatology, Poznan University of Medical Science, Rokietnicka 3 Street, 60-806 Poznan, Poland
| | | | | |
Collapse
|
15
|
López-Cepeda L, Castro JD, Aristizábal-Pachón AF, González-Giraldo Y, Pinzón A, Puentes-Rozo PJ, González J. Modulation of Small RNA Signatures by Astrocytes on Early Neurodegeneration Stages; Implications for Biomarker Discovery. Life (Basel) 2022; 12:1720. [PMID: 36362875 PMCID: PMC9696502 DOI: 10.3390/life12111720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/01/2022] [Accepted: 10/12/2022] [Indexed: 04/04/2024] Open
Abstract
Diagnosis of neurodegenerative disease (NDD) is complex, therefore simpler, less invasive, more accurate biomarkers are needed. small non-coding RNA (sncRNA) dysregulates in NDDs and sncRNA signatures have been explored for the diagnosis of NDDs, however, the performance of previous biomarkers is still better. Astrocyte dysfunction promotes neurodegeneration and thus derived scnRNA signatures could provide a more precise way to identify of changes related to NDD course and pathogenesis, and it could be useful for the dissection of mechanistic insights operating in NDD. Often sncRNA are transported outside the cell by the action of secreted particles such as extracellular vesicles (EV), which protect sncRNA from degradation. Furthermore, EV associated sncRNA can cross the BBB to be found in easier to obtain peripheral samples, EVs also inherit cell-specific surface markers that can be used for the identification of Astrocyte Derived Extracellular Vesicles (ADEVs) in a peripheral sample. By the study of the sncRNA transported in ADEVs it is possible to identify astrocyte specific sncRNA signatures that could show astrocyte dysfunction in a more simpler manner than previous methods. However, sncRNA signatures in ADEV are not a copy of intracellular transcriptome and methodological aspects such as the yield of sncRNA produced in ADEV or the variable amount of ADEV captured after separation protocols must be considered. Here we review the role as signaling molecules of ADEV derived sncRNA dysregulated in conditions associated with risk of neurodegeneration, providing an explanation of why to choose ADEV for the identification of astrocyte-specific transcriptome. Finally, we discuss possible limitations of this approach and the need to improve the detection limits of sncRNA for the use of ADEV derived sncRNA signatures.
Collapse
Affiliation(s)
- Leonardo López-Cepeda
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Juan David Castro
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | | | - Yeimy González-Giraldo
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Andrés Pinzón
- Laboratorio de Bioinformática y Biología de Sistemas, Universidad Nacional de Colombia, Bogotá 111321, Colombia
| | - Pedro J. Puentes-Rozo
- Grupo de Neurociencias del Caribe, Unidad de Neurociencias Cognitivas, Universidad Simón Bolívar, Barranquilla 080002, Colombia
- Grupo de Neurociencias del Caribe, Universidad del Atlántico, Barranquilla 080007, Colombia
| | - Janneth González
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| |
Collapse
|
16
|
Gao W, Zhou J, Gu X, Zhou Y, Wang L, Si N, Fan X, Bian B, Wang H, Zhao H. A multi-network comparative analysis of whole-transcriptome and translatome reveals the effect of high-fat diet on APP/PS1 mice and the intervention with Chinese medicine. Front Nutr 2022; 9:974333. [PMID: 36352898 PMCID: PMC9638104 DOI: 10.3389/fnut.2022.974333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/30/2022] [Indexed: 11/29/2022] Open
Abstract
Different studies on the effects of high-fat diet (HFD) on Alzheimer’s disease (AD) pathology have reported conflicting findings. Our previous studies showed HFD could moderate neuroinflammation and had no significant effect on amyloid-β levels or contextual memory on AD mice. To gain more insights into the involvement of HFD, we performed the whole-transcriptome sequencing and ribosome footprints profiling. Combined with competitive endogenous RNA analysis, the transcriptional regulation mechanism of HFD on AD mice was systematically revealed from RNA level. Mmu-miR-450b-3p and mmu-miR-6540-3p might be involved in regulating the expression of Th and Ddc expression. MiR-551b-5p regulated the expression of a variety of genes including Slc18a2 and Igfbp3. The upregulation of Pcsk9 expression in HFD intervention on AD mice might be closely related to the increase of cholesterol in brain tissues, while Huanglian Jiedu Decoction significantly downregulated the expression of Pcsk9. Our data showed the close connection between the alterations of transcriptome and translatome under the effect of HFD, which emphasized the roles of translational and transcriptional regulation were relatively independent. The profiled molecular responses in current study might be valuable resources for advanced understanding of the mechanisms underlying the effect of HFD on AD.
Collapse
|
17
|
Amelianchik A, Sweetland-Martin L, Norris EH. The effect of dietary fat consumption on Alzheimer's disease pathogenesis in mouse models. Transl Psychiatry 2022; 12:293. [PMID: 35869065 PMCID: PMC9307654 DOI: 10.1038/s41398-022-02067-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer's disease (AD) is a fatal cognitive disorder with proteinaceous brain deposits, neuroinflammation, cerebrovascular dysfunction, and extensive neuronal loss over time. AD is a multifactorial disease, and lifestyle factors, including diet, are likely associated with the development of AD pathology. Since obesity and diabetes are recognized as risk factors for AD, it might be predicted that a high-fat diet (HFD) would worsen AD pathology. However, modeling HFD-induced obesity in AD animal models has yielded inconclusive results. Some studies report a deleterious effect of HFD on Aβ accumulation, neuroinflammation, and cognitive function, while others report that HFD worsens memory without affecting AD brain pathology. Moreover, several studies report no major effect of HFD on AD-related phenotypes in mice, while other studies show that HFD might, in fact, be protective. The lack of a clear association between dietary fat consumption and AD-related pathology and cognitive function in AD mouse models might be explained by experimental variations, including AD mouse model, sex and age of the animals, composition of the HFD, and timeline of HFD consumption. In this review, we summarize recent studies that aimed at elucidating the effect of HFD-induced obesity on AD-related pathology in mice and provide an overview of the factors that may have contributed to the results reported in these studies. Based on the heterogeneity of these animal model studies and given that the human population itself is quite disparate, it is likely that people will benefit most from individualized nutritional plans based on their medical history and clinical profiles.
Collapse
Affiliation(s)
- Anna Amelianchik
- Patricia and John Rosenwald Laboratory of Neurobiology & Genetics, The Rockefeller University, 1230 York Avenue, New York, NY, USA
| | - Lauren Sweetland-Martin
- Patricia and John Rosenwald Laboratory of Neurobiology & Genetics, The Rockefeller University, 1230 York Avenue, New York, NY, USA
| | - Erin H Norris
- Patricia and John Rosenwald Laboratory of Neurobiology & Genetics, The Rockefeller University, 1230 York Avenue, New York, NY, USA.
| |
Collapse
|
18
|
Liu R, Bai L, Liu M, Wang R, Wu Y, Li Q, Ba Y, Zhang H, Zhou G, Yu F, Huang H. Combined exposure of lead and high-fat diet enhanced cognitive decline via interacting with CREB-BDNF signaling in male rats. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 304:119200. [PMID: 35364187 DOI: 10.1016/j.envpol.2022.119200] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/03/2022] [Accepted: 03/21/2022] [Indexed: 06/14/2023]
Abstract
The health risks to populations induced by lead (Pb) and high-fat diets (HFD) have become a global public health problem. Pb and HFD often co-exist and are co-occurring risk factors for cognitive impairment. This study investigates effect of combined Pb and HFD on cognitive function, and explores the underlying mechanisms in terms of regulatory components of synaptic plasticity and insulin signaling pathway. We showed that the co-exposure of Pb and HFD further increased blood Pb levels, caused body weight loss and dyslipidemia. The results from Morris water maze (MWM) test and Nissl staining disclosed that Pb and HFD each contributed to cognitive deficits and neuronal damage and combined exposure enhanced this toxic injury. Pb and HFD decreased the levels of synapsin-1, GAP-43 and PSD-95 protein related to synaptic properties and SIRT1, NMDARs, phosphorylated CREB and BDNF related to synaptic plasticity regulatory, and these decreases was greater when combined exposure. Additionally, we revealed that Pb and HFD promoted IRS-1 phosphorylation and subsequently reduced downstream PI3K-Akt kinases phosphorylation in hippocampus and cortex of rats, and this process was aggravated when co-exposure. Collectively, our data suggested that combined exposure of Pb and HFD enhanced cognitive deficits, pointing to additive effects in rats than the individual stress effects related to multiple signaling pathways with CREB-BDNF signaling as the hub. This study emphasizes the need to evaluate the effects of mixed exposures on brain function in realistic environment and to better inform prevention of neurological disorders via modulating central pathway, such as CREB/BDNF signaling.
Collapse
Affiliation(s)
- Rundong Liu
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Lin Bai
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Mengchen Liu
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Ruike Wang
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Yingying Wu
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Qiong Li
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Yue Ba
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Huizhen Zhang
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Guoyu Zhou
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Fangfang Yu
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Hui Huang
- Department of Environmental Health &Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
19
|
Nehra G, Bauer B, Hartz AMS. Blood-brain barrier leakage in Alzheimer's disease: From discovery to clinical relevance. Pharmacol Ther 2022; 234:108119. [PMID: 35108575 PMCID: PMC9107516 DOI: 10.1016/j.pharmthera.2022.108119] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia. AD brain pathology starts decades before the onset of clinical symptoms. One early pathological hallmark is blood-brain barrier dysfunction characterized by barrier leakage and associated with cognitive decline. In this review, we summarize the existing literature on the extent and clinical relevance of barrier leakage in AD. First, we focus on AD animal models and their susceptibility to barrier leakage based on age and genetic background. Second, we re-examine barrier dysfunction in clinical and postmortem studies, summarize changes that lead to barrier leakage in patients and highlight the clinical relevance of barrier leakage in AD. Third, we summarize signaling mechanisms that link barrier leakage to neurodegeneration and cognitive decline in AD. Finally, we discuss clinical relevance and potential therapeutic strategies and provide future perspectives on investigating barrier leakage in AD. Identifying mechanistic steps underlying barrier leakage has the potential to unravel new targets that can be used to develop novel therapeutic strategies to repair barrier leakage and slow cognitive decline in AD and AD-related dementias.
Collapse
Affiliation(s)
- Geetika Nehra
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Bjoern Bauer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, USA; Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
20
|
Wang J, Wang K, Liu T, Wang L, Suo D, Xie Y, Funahashi S, Wu J, Pei G. Abnormal Dynamic Functional Networks in Subjective Cognitive Decline and Alzheimer's Disease. Front Comput Neurosci 2022; 16:885126. [PMID: 35586480 PMCID: PMC9108158 DOI: 10.3389/fncom.2022.885126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Subjective cognitive decline (SCD) is considered to be the preclinical stage of Alzheimer's disease (AD) and has the potential for the early diagnosis and intervention of AD. It was implicated that CSF-tau, which increases very early in the disease process in AD, has a high sensitivity and specificity to differentiate AD from normal aging, and the highly connected brain regions behaved more tau burden in patients with AD. Thus, a highly connected state measured by dynamic functional connectivity may serve as the early changes of AD. In this study, forty-five normal controls (NC), thirty-six individuals with SCD, and thirty-five patients with AD were enrolled to obtain the resting-state functional magnetic resonance imaging scanning. Sliding windows, Pearson correlation, and clustering analysis were combined to investigate the different levels of information transformation states. Three states, namely, the low state, the middle state, and the high state, were characterized based on the strength of functional connectivity between each pair of brain regions. For the global dynamic functional connectivity analysis, statistically significant differences were found among groups in the three states, and the functional connectivity in the middle state was positively correlated with cognitive scales. Furthermore, the whole brain was parcellated into four networks, namely, default mode network (DMN), cognitive control network (CCN), sensorimotor network (SMN), and occipital-cerebellum network (OCN). For the local network analysis, statistically significant differences in CCN for low state and SMN for middle state and high state were found in normal controls and patients with AD. Meanwhile, the differences were also found in normal controls and individuals with SCD. In addition, the functional connectivity in SMN for high state was positively correlated with cognitive scales. Converging results showed the changes in dynamic functional states in individuals with SCD and patients with AD. In addition, the changes were mainly in the high strength of the functional connectivity state.
Collapse
Affiliation(s)
- Jue Wang
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Kexin Wang
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Tiantian Liu
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Li Wang
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Dingjie Suo
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Yunyan Xie
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shintaro Funahashi
- Kokoro Research Center, Kyoto University, Kyoto, Japan
- Laboratory of Cognitive Brain Science, Department of Cognitive and Behavioral Sciences, Graduate School of Human and Environmental Studies, Kyoto University, Kyoto, Japan
| | - Jinglong Wu
- Research Center for Medical Artificial Intelligence, Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, China
- *Correspondence: Jinglong Wu
| | - Guangying Pei
- School of Life Science, Beijing Institute of Technology, Beijing, China
- Guangying Pei
| |
Collapse
|
21
|
Xu CJ, Li MQ, Li-Zhao, Chen WG, Wang JL. Short-term high-fat diet favors the appearances of apoptosis and gliosis by activation of ERK1/2/p38MAPK pathways in brain. Aging (Albany NY) 2021; 13:23133-23148. [PMID: 34620734 PMCID: PMC8544319 DOI: 10.18632/aging.203607] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/07/2021] [Indexed: 01/26/2023]
Abstract
High-fat diet (HFD) has been associated with neuroinflammation and apoptosis in distinct brain regions. To explore the effect of short-term (7, 14 and 21 days) high-fat overfeeding on apoptosis, inflammatory signaling proteins, APP changes and glial cell activities in cerebral cortex and cerebellum. Mice were fed with HFD for different lengths (up to 21 days) and after each time body weights of mice was tested, then the apoptotic proteins, IL-1β, APP, BACE1and MAPKs, Akt and NF-κB signaling activity were evaluated by western blots. Results demonstrate that short period of high-fat overnutrition significantly promotes apoptosis, APP expression at day 21 of cerebral cortex and at day 7 of cerebellum compared to chow diet. In addition, increased GFAP+astrocytes, Iba-1+microglia and IL-1β 30 were observed in cerebral cortex after 21 days HFD, but no changes for 7 days overfeeding of cerebellum. Serendipitously, ERK1/2 pathway was activated both in cerebral cortex and cerebellum for different time course of HFD. Furthermore, increased phospho-p38 MAPK level was observed in cerebellum only. In consistent with in vivo results, SH-SY5Y cells treatment with cholesterol (50 μM, 100 μM) for 48 h culture in vitro demonstrated that pro-apoptotic proteins were enhanced as well. In brief, short-term HFD consumption increases sensitivity to apoptosis, APP and IL-1β production as well as gliosis in cerebral cortex and cerebellum, which may be related to enhancement of ERK1/2 and p38 MAPK activation.
Collapse
Affiliation(s)
- Chao-Jin Xu
- Department of Histology and Embryology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Mei-Qi Li
- School of 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Li-Zhao
- School of 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Wei-Guang Chen
- Department of Histology and Embryology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Jun-Ling Wang
- Center for Reproductive Medicine, Affiliated Hospital 1 of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| |
Collapse
|
22
|
Yanguas-Casás N, Torres C, Crespo-Castrillo A, Diaz-Pacheco S, Healy K, Stanton C, Chowen JA, Garcia-Segura LM, Arevalo MA, Cryan JF, de Ceballos ML. High-fat diet alters stress behavior, inflammatory parameters and gut microbiota in Tg APP mice in a sex-specific manner. Neurobiol Dis 2021; 159:105495. [PMID: 34478848 DOI: 10.1016/j.nbd.2021.105495] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/13/2021] [Accepted: 08/27/2021] [Indexed: 12/12/2022] Open
Abstract
Long-term high-fat diet (HFD) consumption commonly leads to obesity, a major health concern of western societies and a risk factor for Alzheimer's disease (AD). Both conditions present glial activation and inflammation and show sex differences in their incidence, clinical manifestation, and disease course. HFD intake has an important impact on gut microbiota, the bacteria present in the gut, and microbiota dysbiosis is associated with inflammation and certain mental disorders such as anxiety. In this study, we have analyzed the effects of a prolonged (18 weeks, starting at 7 months of age) HFD on male and female mice, both wild type (WT) and TgAPP mice, a model for AD, investigating the behavioral profile, gut microbiota composition and inflammatory/phagocytosis-related gene expression in hippocampus. In the open-field test, no overt differences in motor activity were observed between male and female or WT and TgAPP mice on a low-fat diet (LFD). However, HFD induced anxiety, as judged by decreased motor activity and increased time in the margins in the open-field, and a trend towards increased immobility time in the tail suspension test, with increased defecation. Intriguingly, female TgAPP mice on HFD showed less immobility and defecation compared to female WT mice on HFD. HFD induced dysbiosis of gut microbiota, resulting in reduced microbiota diversity and abundance compared with LFD fed mice, with some significant differences due to sex and little effect of genotype. Gene expression of pro-inflammatory/phagocytic markers in the hippocampus were not different between male and female WT mice, and in TgAPP mice of both sexes, some cytokines (IL-6 and IFNγ) were higher than in WT mice on LFD, more so in female TgAPP (IL-6). HFD induced few alterations in mRNA expression of inflammatory/phagocytosis-related genes in male mice, whether WT (IL-1β, MHCII), or TgAPP (IL-6). However, in female TgAPP, altered gene expression returned towards control levels following prolonged HFD (IL-6, IL-12β, TNFα, CD36, IRAK4, PYRY6). In summary, we demonstrate that HFD induces anxiogenic symptoms, marked alterations in gut microbiota, and increased expression of inflammatory genes, except for female TgAPP that appear to be resistant to the diet effects. Lifestyle interventions should be introduced to prevent AD onset or exacerbation by reducing inflammation and its associated symptoms; however, our results suggest that the eventual goal of developing prevention and treatment strategies should take sex into consideration.
Collapse
Affiliation(s)
- Natalia Yanguas-Casás
- Cajal Institute, CSIC, 28002 Madrid, Spain; Centre for Biomedical Network Research for Frailty and Healthy Ageing (CIBERFES) Instituto de Salud Carlos III, Madrid, Spain; Lymphoma Research Group, Medical Oncology Department, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, Majadahonda, Madrid, Spain
| | - Cristina Torres
- Dept Anatomy & Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland; Universitat Rovira i Virgili, Biochemistry and Biotechnology Department, 43007 Tarragona, Spain
| | | | | | - Kiera Healy
- Dept Anatomy & Neuroscience, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Catherine Stanton
- Dept Anatomy & Neuroscience, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, 28009 Madrid, Spain; Centre for Biomedical Network Research for Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; The Madrid Institute for the advanced study of Food (IMDEA de Alimentación), Madrid, Spain
| | - Luis M Garcia-Segura
- Cajal Institute, CSIC, 28002 Madrid, Spain; Centre for Biomedical Network Research for Frailty and Healthy Ageing (CIBERFES) Instituto de Salud Carlos III, Madrid, Spain
| | - Maria Angeles Arevalo
- Cajal Institute, CSIC, 28002 Madrid, Spain; Centre for Biomedical Network Research for Frailty and Healthy Ageing (CIBERFES) Instituto de Salud Carlos III, Madrid, Spain
| | - John F Cryan
- Dept Anatomy & Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | |
Collapse
|
23
|
Mazzei G, Ikegami R, Abolhassani N, Haruyama N, Sakumi K, Saito T, Saido TC, Nakabeppu Y. A high-fat diet exacerbates the Alzheimer's disease pathology in the hippocampus of the App NL-F/NL-F knock-in mouse model. Aging Cell 2021; 20:e13429. [PMID: 34245097 PMCID: PMC8373331 DOI: 10.1111/acel.13429] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 06/01/2021] [Accepted: 06/15/2021] [Indexed: 12/14/2022] Open
Abstract
Insulin resistance and diabetes mellitus are major risk factors for Alzheimer's disease (AD), and studies with transgenic mouse models of AD have provided supportive evidence with some controversies. To overcome potential artifacts derived from transgenes, we used a knock‐in mouse model, AppNL−F/NL−F, which accumulates Aβ plaques from 6 months of age and shows mild cognitive impairment at 18 months of age, without the overproduction of APP. In the present study, 6‐month‐old male AppNL−F/NL−F and wild‐type mice were fed a regular or high‐fat diet (HFD) for 12 months. HFD treatment caused obesity and impaired glucose tolerance (i.e., T2DM conditions) in both wild‐type and AppNL−F/NL−F mice, but only the latter animals exhibited an impaired cognitive function accompanied by marked increases in both Aβ deposition and microgliosis as well as insulin resistance in the hippocampus. Furthermore, HFD‐fed AppNL−F/NL−F mice exhibited a significant decrease in volume of the granule cell layer in the dentate gyrus and an increased accumulation of 8‐oxoguanine, an oxidized guanine base, in the nuclei of granule cells. Gene expression profiling by microarrays revealed that the populations of the cell types in hippocampus were not significantly different between the two mouse lines, regardless of the diet. In addition, HFD treatment decreased the expression of the Aβ binding protein transthyretin (TTR) in AppNL−F/NL−F mice, suggesting that the depletion of TTR underlies the increased Aβ deposition in the hippocampus of HFD‐fed AppNL−F/NL−F mice.
Collapse
Affiliation(s)
- Guianfranco Mazzei
- Division of Neurofunctional Genomics Department of Immunobiology and Neuroscience Medical Institute of Bioregulation Kyushu University Fukuoka Japan
| | - Ryohei Ikegami
- Division of Neurofunctional Genomics Department of Immunobiology and Neuroscience Medical Institute of Bioregulation Kyushu University Fukuoka Japan
| | - Nona Abolhassani
- Division of Neurofunctional Genomics Department of Immunobiology and Neuroscience Medical Institute of Bioregulation Kyushu University Fukuoka Japan
| | - Naoki Haruyama
- Division of Neurofunctional Genomics Department of Immunobiology and Neuroscience Medical Institute of Bioregulation Kyushu University Fukuoka Japan
| | - Kunihiko Sakumi
- Division of Neurofunctional Genomics Department of Immunobiology and Neuroscience Medical Institute of Bioregulation Kyushu University Fukuoka Japan
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience RIKEN Center for Brain Science Saitama Japan
- Department of Neurocognitive Science Institute of Brain Science Nagoya City University Graduate School of Medical Sciences Nagoya Japan
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience RIKEN Center for Brain Science Saitama Japan
| | - Yusaku Nakabeppu
- Division of Neurofunctional Genomics Department of Immunobiology and Neuroscience Medical Institute of Bioregulation Kyushu University Fukuoka Japan
| |
Collapse
|
24
|
Fan X, Liu B, Zhou J, Gu X, Zhou Y, Yang Y, Guo F, Wei X, Wang H, Si N, Yang J, Bian B, Zhao H. High-Fat Diet Alleviates Neuroinflammation and Metabolic Disorders of APP/PS1 Mice and the Intervention With Chinese Medicine. Front Aging Neurosci 2021; 13:658376. [PMID: 34168550 PMCID: PMC8217439 DOI: 10.3389/fnagi.2021.658376] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/06/2021] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease caused by the complex interaction of multiple mechanisms. Recent studies examining the effect of high-fat diet (HFD) on the AD phenotype have demonstrated a significant influence on both inflammation and cognition. However, different studies on the effect of high-fat diet on AD pathology have reported conflicting conclusions. To explore the involvement of HFD in AD, we investigated phenotypic and metabolic changes in an AD mouse model in response to HFD. The results indicated there was no significant effect on Aβ levels or contextual memory due to HFD treatment. Of note, HFD did moderate neuroinflammation, despite spurring inflammation and increasing cholesterol levels in the periphery. In addition, diet affected gut microbiota symbiosis, altering the production of bacterial metabolites. HFD created a favorable microenvironment for bile acid alteration and arachidonic acid metabolism in APP/PS1 mice, which may be related to the observed improvement in LXR/PPAR expression. Our previous research demonstrated that Huanglian Jiedu decoction (HLJDD) significantly ameliorated impaired learning and memory. Furthermore, HLJDD may globally suppress inflammation and lipid accumulation to relieve cognitive impairment after HFD intervention. It was difficult to define the effect of HFD on AD progression because the results were influenced by confounding factors and biases. Although there was still obvious damage in AD mice treated with HFD, there was no deterioration and there was even a slight remission of neuroinflammation. Moreover, HLJDD represents a potential AD drug based on its anti-inflammatory and lipid-lowering effects.
Collapse
Affiliation(s)
- Xiaorui Fan
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China.,Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bin Liu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Junyi Zhou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinru Gu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanyan Zhou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yifei Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Feifei Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaolu Wei
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongjie Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Nan Si
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jian Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baolin Bian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haiyu Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
25
|
Amelianchik A, Merkel J, Palanisamy P, Kaneki S, Hyatt E, Norris EH. The protective effect of early dietary fat consumption on Alzheimer's disease-related pathology and cognitive function in mice. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12173. [PMID: 34084889 PMCID: PMC8144936 DOI: 10.1002/trc2.12173] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 03/24/2021] [Accepted: 04/05/2021] [Indexed: 11/12/2022]
Abstract
INTRODUCTION It has been suggested that obesity may influence Alzheimer's disease (AD) pathogenesis, yet the numerous publications on this topic have inconsistent results and conclusions. METHODS Our study examined the effect of varying the timing of high-fat diet (HFD) consumption on AD-related pathology and cognition in transgenic Tg6799 AD mice. RESULTS HFD feeding starting at or before 3 months of age, prior to severe AD pathology, had protective effects in AD mice: reduced extracellular amyloid beta (Aβ) deposition, decreased fibrinogen extravasation into the brain parenchyma, and improved cognitive function. However, delaying HFD consumption until 6 months of age, when AD pathology is ubiquitous, reduced these protective effects in AD mice. DISCUSSION Overall, we demonstrate that the timeline of HFD consumption may play an important role in how dietary fats affect AD pathogenesis and cognitive function.
Collapse
Affiliation(s)
- Anna Amelianchik
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkUSA
| | - Jonathan Merkel
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkUSA
- Paul Flechsig Institute of Brain ResearchLeipzig UniversityLeipzigGermany
| | - Premkumar Palanisamy
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkUSA
| | - Shigeru Kaneki
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkUSA
| | - Emily Hyatt
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkUSA
| | - Erin H. Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkUSA
| |
Collapse
|
26
|
Nakai T, Yamada K, Mizoguchi H. Alzheimer's Disease Animal Models: Elucidation of Biomarkers and Therapeutic Approaches for Cognitive Impairment. Int J Mol Sci 2021; 22:5549. [PMID: 34074018 PMCID: PMC8197360 DOI: 10.3390/ijms22115549] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is an age-related and progressive neurodegenerative disorder. It is widely accepted that AD is mainly caused by the accumulation of extracellular amyloid β (Aβ) and intracellular neurofibrillary tau tangles. Aβ begins to accumulate years before the onset of cognitive impairment, suggesting that the benefit of currently available interventions would be greater if they were initiated in the early phases of AD. To understand the mechanisms of AD pathogenesis, various transgenic mouse models with an accelerated accumulation of Aβ and tau tangles have been developed. However, none of these models exhibit all pathologies present in human AD. To overcome these undesirable phenotypes, APP knock-in mice, which were presented with touchscreen-based tasks, were developed to better evaluate the efficacy of candidate therapeutics in mouse models of early-stage AD. This review assesses several AD mouse models from the aspect of biomarkers and cognitive impairment and discusses their potential as tools to provide novel AD therapeutic approaches.
Collapse
Affiliation(s)
- Tsuyoshi Nakai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan; (T.N.); (K.Y.)
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan; (T.N.); (K.Y.)
| | - Hiroyuki Mizoguchi
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan; (T.N.); (K.Y.)
- Medical Interactive Research and Academia Industry Collaboration Center, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
| |
Collapse
|
27
|
Nyúl-Tóth Á, Tarantini S, DelFavero J, Yan F, Balasubramanian P, Yabluchanskiy A, Ahire C, Kiss T, Csipo T, Lipecz A, Farkas AE, Wilhelm I, Krizbai IA, Tang Q, Csiszar A, Ungvari Z. Demonstration of age-related blood-brain barrier disruption and cerebromicrovascular rarefaction in mice by longitudinal intravital two-photon microscopy and optical coherence tomography. Am J Physiol Heart Circ Physiol 2021; 320:H1370-H1392. [PMID: 33543687 PMCID: PMC8260380 DOI: 10.1152/ajpheart.00709.2020] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 01/21/2021] [Accepted: 01/21/2021] [Indexed: 12/25/2022]
Abstract
Age-related blood-brain barrier (BBB) disruption and cerebromicrovascular rarefaction contribute importantly to the pathogenesis of both vascular cognitive impairment and dementia (VCID) and Alzheimer's disease (AD). Recent advances in geroscience research enable development of novel interventions to reverse age-related alterations of the cerebral microcirculation for prevention of VCID and AD. To facilitate this research, there is an urgent need for sensitive and easy-to-adapt imaging methods that enable longitudinal assessment of changes in BBB permeability and brain capillarization in aged mice and that could be used in vivo to evaluate treatment efficiency. To enable longitudinal assessment of changes in BBB permeability in aged mice equipped with a chronic cranial window, we adapted and optimized two different intravital two-photon imaging approaches. By assessing relative fluorescence changes over the baseline within a volume of brain tissue, after qualitative image subtraction of the brain microvasculature, we confirmed that, in 24-mo-old C57BL/6J mice, cumulative permeability of the microvessels to fluorescent tracers of different molecular masses (0.3 to 40 kDa) is significantly increased compared with that of 5-mo-old mice. Real-time recording of vessel cross-sections showed that apparent solute permeability of single microvessels is significantly increased in aged mice vs. young mice. Cortical capillary density, assessed both by intravital two-photon microscopy and optical coherence tomography was also decreased in aged mice vs. young mice. The presented methods have been optimized for longitudinal (over the period of 36 wk) in vivo assessment of cerebromicrovascular health in preclinical geroscience research.NEW & NOTEWORTHY Methods are presented for longitudinal detection of age-related increase in blood-brain barrier permeability and microvascular rarefaction in the mouse cerebral cortex by intravital two-photon microscopy and optical coherence tomography.
Collapse
Affiliation(s)
- Ádám Nyúl-Tóth
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center For Geroscience and Healthy Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- International Training Program in Geroscience, Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center For Geroscience and Healthy Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Jordan DelFavero
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center For Geroscience and Healthy Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Feng Yan
- Stephenson School of Biomedical Engineering, Gallogly College of Engineering, The University of Oklahoma, Norman, Oklahoma
| | - Priya Balasubramanian
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center For Geroscience and Healthy Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center For Geroscience and Healthy Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Chetan Ahire
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center For Geroscience and Healthy Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Tamas Kiss
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center For Geroscience and Healthy Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- International Training Program in Geroscience, Theoretical Medicine Doctoral School/Departments of Medical Physics and Informatics and Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
| | - Tamas Csipo
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center For Geroscience and Healthy Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Agnes Lipecz
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center For Geroscience and Healthy Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Attila E Farkas
- International Training Program in Geroscience, Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
| | - Imola Wilhelm
- International Training Program in Geroscience, Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, Arad, Romania
| | - István A Krizbai
- International Training Program in Geroscience, Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, Arad, Romania
| | - Qinggong Tang
- Stephenson School of Biomedical Engineering, Gallogly College of Engineering, The University of Oklahoma, Norman, Oklahoma
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center For Geroscience and Healthy Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Theoretical Medicine Doctoral School/Departments of Medical Physics and Informatics and Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center For Geroscience and Healthy Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- International Training Program in Geroscience, Theoretical Medicine Doctoral School/Departments of Medical Physics and Informatics and Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
28
|
Leikin-Frenkel A, Liraz-Zaltsman S, Hollander KS, Atrakchi D, Ravid O, Rand D, Kandel-Kfir M, Israelov H, Cohen H, Kamari Y, Shaish A, Harats D, Schnaider-Beeri M, Cooper I. Dietary alpha linolenic acid in pregnant mice and during weaning increases brain docosahexaenoic acid and improves recognition memory in the offspring. J Nutr Biochem 2021; 91:108597. [PMID: 33545323 DOI: 10.1016/j.jnutbio.2021.108597] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 12/22/2020] [Accepted: 01/05/2021] [Indexed: 11/17/2022]
Abstract
Docosahexaenoic acid (DHA) is critical for normal brain development and function. DHA is in danger of being significantly reduced in the human food supply, and the question of whether its metabolic precursor, the essential n-3 alpha linolenic acid (ALA) during pregnancy, can support fetal brain DHA levels for optimal neurodevelopment, is fundamental. Female mice were fed either ALA-enriched or Control diet during pregnancy and lactation. The direct effect of maternal dietary ALA on lipids was analyzed in liver, red blood cells, brain and brain vasculature, together with genes of fatty acid metabolism and transport in three-week-old offspring. The long-term effect of maternal dietary ALA on brain fatty acids and memory was studied in 19-week-old offspring. Three-week-old ALA offspring showed higher levels of n-3 fatty acids in liver, red blood cell, blood-brain barrier (BBB) vasculature and brain parenchyma, DHA enrichment in brain phospholipids and higher gene and protein expression of the DHA transporter, major facilitator superfamily domain containing 2a, compared to Controls. 19-week-old ALA offspring showed higher brain DHA levels and better memory performance than Controls. The increased brain DHA levels induced by maternal dietary ALA during pregnancy-lactation, together with the up-regulated levels of major facilitator superfamily domain containing 2a, may indicate a mode for greater DHA uptake with long-term impact on better memory in ALA offspring.
Collapse
Affiliation(s)
- Alicia Leikin-Frenkel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Bert Strassburger Lipid Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Sigal Liraz-Zaltsman
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel-Hashomer, Israel
| | | | - Dana Atrakchi
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Orly Ravid
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Daniel Rand
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Michal Kandel-Kfir
- Bert Strassburger Lipid Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Hila Israelov
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Hofit Cohen
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Bert Strassburger Lipid Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Yehuda Kamari
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Bert Strassburger Lipid Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Aviv Shaish
- Bert Strassburger Lipid Center, Sheba Medical Center, Tel-Hashomer, Israel; Achva Academic College, Israel
| | - Dror Harats
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Bert Strassburger Lipid Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Michal Schnaider-Beeri
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel-Hashomer, Israel; School of Psychology, Interdisciplinary Center (IDC) Herzliya, Herzliya, Israel; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Itzik Cooper
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel-Hashomer, Israel; School of Psychology, Interdisciplinary Center (IDC) Herzliya, Herzliya, Israel; The Nehemia Rubin Excellence in Biomedical Research - The TELEM Program, Sheba Medical Center, Tel-Hashomer, Israel.
| |
Collapse
|
29
|
de Bem AF, Krolow R, Farias HR, de Rezende VL, Gelain DP, Moreira JCF, Duarte JMDN, de Oliveira J. Animal Models of Metabolic Disorders in the Study of Neurodegenerative Diseases: An Overview. Front Neurosci 2021; 14:604150. [PMID: 33536868 PMCID: PMC7848140 DOI: 10.3389/fnins.2020.604150] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/24/2020] [Indexed: 12/21/2022] Open
Abstract
The incidence of metabolic disorders, as well as of neurodegenerative diseases—mainly the sporadic forms of Alzheimer’s and Parkinson’s disease—are increasing worldwide. Notably, obesity, diabetes, and hypercholesterolemia have been indicated as early risk factors for sporadic forms of Alzheimer’s and Parkinson’s disease. These conditions share a range of molecular and cellular features, including protein aggregation, oxidative stress, neuroinflammation, and blood-brain barrier dysfunction, all of which contribute to neuronal death and cognitive impairment. Rodent models of obesity, diabetes, and hypercholesterolemia exhibit all the hallmarks of these degenerative diseases, and represent an interesting approach to the study of the phenotypic features and pathogenic mechanisms of neurodegenerative disorders. We review the main pathological aspects of Alzheimer’s and Parkinson’s disease as summarized in rodent models of obesity, diabetes, and hypercholesterolemia.
Collapse
Affiliation(s)
- Andreza Fabro de Bem
- Department of Physiological Sciences, Institute of Biology, University of Brasilia, Brazilia, Brazil
| | - Rachel Krolow
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Hémelin Resende Farias
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Victória Linden de Rezende
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Daniel Pens Gelain
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - José Cláudio Fonseca Moreira
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - João Miguel das Neves Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Jade de Oliveira
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
30
|
Shabir O, Moll TA, Matuszyk MM, Eyre B, Dake MD, Berwick J, Francis SE. Preclinical models of disease and multimorbidity with focus upon cardiovascular disease and dementia. Mech Ageing Dev 2020; 192:111361. [PMID: 32998028 DOI: 10.1016/j.mad.2020.111361] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/28/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022]
|
31
|
Bomba M, Granzotto A, Castelli V, Onofrj M, Lattanzio R, Cimini A, Sensi SL. Exenatide Reverts the High-Fat-Diet-Induced Impairment of BDNF Signaling and Inflammatory Response in an Animal Model of Alzheimer's Disease. J Alzheimers Dis 2020; 70:793-810. [PMID: 31256135 DOI: 10.3233/jad-190237] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial condition in which, along with amyloid-β (Aβ) and tau-related pathology, the synergistic activity of co-morbidity factors promote the onset and progression of the disease. Epidemiological evidence indicates that glucose intolerance, deficits in insulin secretion, or type-2 diabetes mellitus (T2DM) participate in increasing cognitive impairment or dementia risk. Insulin plays a pivotal role in the process as the hormone critically regulates brain functioning. GLP-1, the glucagon-like peptide 1, facilitates insulin signaling, regulates glucose homeostasis, and modulates synaptic plasticity. Exenatide is a synthetic GLP-1 analog employed in T2DM. However, exenatide has also been shown to affect the signaling of the brain-derived neurotrophic factor (BDNF), synaptic plasticity, and cognitive performances in animal models. In this study, we tested whether exenatide exerts neuroprotection in a preclinical AD model set to mimic the clinical complexity of the human disease. We investigated the effects of exenatide treatment in 3xTg-AD mice challenged with a high-fat diet (HFD). Endpoints of the study were variations in systemic metabolism, insulin and neurotrophic signaling, neuroinflammation, Aβ and tau pathology, and cognitive performances. Results of the study indicate that exenatide reverts the adverse changes of BDNF signaling and the neuroinflammation status of 3xTg-AD mice undergoing HFD without affecting systemic metabolism or promoting changes in cognitive performances.
Collapse
Affiliation(s)
- Manuela Bomba
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, University G. d'Annunzio of Chieti-Pescara, Italy.,Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Italy
| | - Alberto Granzotto
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, University G. d'Annunzio of Chieti-Pescara, Italy.,Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy
| | - Marco Onofrj
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, University G. d'Annunzio of Chieti-Pescara, Italy.,Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Italy
| | - Rossano Lattanzio
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, University G. d'Annunzio of Chieti-Pescara, Italy.,Department of Medical, Oral, and Biotechnological Sciences, University G. d'Annunzio of Chieti-Pescara, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, PA, USA.,National Institute for Nuclear Physics (INFN), Gran Sasso National Laboratory (LNGS), Assergi, Italy
| | - Stefano L Sensi
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, University G. d'Annunzio of Chieti-Pescara, Italy.,Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Italy.,Departments of Neurology and Pharmacology, Institute for Mind Impairments and Neurological Disorders - iMIND, University of California - Irvine, Irvine, CA, USA
| |
Collapse
|
32
|
IL-6 Trans-Signaling in the Brain Influences the Metabolic Phenotype of the 3xTg-AD Mouse Model of Alzheimer's Disease. Cells 2020; 9:cells9071605. [PMID: 32630818 PMCID: PMC7407279 DOI: 10.3390/cells9071605] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that causes the most prevalent dementia in the elderly people. Obesity and insulin resistance, which may cause major health problems per se, are risk factors for AD, and cytokines such as interleukin-6 (IL-6) have a role in these conditions. IL-6 can signal either through a membrane receptor or by trans-signaling, which can be inhibited by the soluble form of the co-receptor gp130 (sgp130). We have addressed the possibility that blocking IL-6 trans-signaling in the brain could have an effect in the triple transgenic 3xTg-AD mouse model of AD and/or in obesity progression, by crossing 3xTg-AD mice with GFAP-sgp130Fc mice. To serve as control groups, GFAP-sgp130Fc mice were also crossed with C57BL/6JOlaHsd mice. Seventeen-month-old mice were fed a control diet (18% kcal from fat) and a high-fat diet (HFD; 58.4% kcal from fat). In our experimental conditions, the 3xTg-AD model showed a mild amyloid phenotype, which nevertheless altered the control of body weight and related endocrine and metabolic factors, suggestive of a hypermetabolic state. The inhibition of IL-6 trans-signaling modulated some of these traits in both 3xTg-AD and control mice, particularly during HFD, and in a sex-dependent manner. These experiments provide evidence of IL-6 trans-signaling playing a role in the CNS of a mouse model of AD.
Collapse
|
33
|
Bracko O, Vinarcsik LK, Cruz Hernández JC, Ruiz-Uribe NE, Haft-Javaherian M, Falkenhain K, Ramanauskaite EM, Ali M, Mohapatra A, Swallow MA, Njiru BN, Muse V, Michelucci PE, Nishimura N, Schaffer CB. High fat diet worsens Alzheimer's disease-related behavioral abnormalities and neuropathology in APP/PS1 mice, but not by synergistically decreasing cerebral blood flow. Sci Rep 2020; 10:9884. [PMID: 32555372 PMCID: PMC7303150 DOI: 10.1038/s41598-020-65908-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/07/2020] [Indexed: 12/18/2022] Open
Abstract
Obesity is linked to increased risk for and severity of Alzheimer's disease (AD). Cerebral blood flow (CBF) reductions are an early feature of AD and are also linked to obesity. We recently showed that non-flowing capillaries, caused by adhered neutrophils, contribute to CBF reduction in mouse models of AD. Because obesity could exacerbate the vascular inflammation likely underlying this neutrophil adhesion, we tested links between obesity and AD by feeding APP/PS1 mice a high fat diet (Hfd) and evaluating behavioral, physiological, and pathological changes. We found trends toward poorer memory performance in APP/PS1 mice fed a Hfd, impaired social interactions with either APP/PS1 genotype or a Hfd, and synergistic impairment of sensory-motor function in APP/PS1 mice fed a Hfd. The Hfd led to increases in amyloid-beta monomers and plaques in APP/PS1 mice, as well as increased brain inflammation. These results agree with previous reports showing obesity exacerbates AD-related pathology and symptoms in mice. We used a crowd-sourced, citizen science approach to analyze imaging data to determine the impact of the APP/PS1 genotype and a Hfd on capillary stalling and CBF. Surprisingly, we did not see an increase in the number of non-flowing capillaries or a worsening of the CBF deficit in APP/PS1 mice fed a Hfd as compared to controls, suggesting that capillary stalling is not a mechanistic link between a Hfd and increased severity of AD in mice. Reducing capillary stalling by blocking neutrophil adhesion improved CBF and short-term memory function in APP/PS1 mice, even when fed a Hfd.
Collapse
Affiliation(s)
- Oliver Bracko
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Lindsay K Vinarcsik
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | | | - Nancy E Ruiz-Uribe
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | | | - Kaja Falkenhain
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | | | - Muhammad Ali
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Aditi Mohapatra
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Madisen A Swallow
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Brendah N Njiru
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Victorine Muse
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | | | - Nozomi Nishimura
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Chris B Schaffer
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
34
|
Antioxidant Effects of Walnut ( Juglans regia L.) Kernel and Walnut Septum Extract in a D-Galactose-Induced Aging Model and in Naturally Aged Rats. Antioxidants (Basel) 2020; 9:antiox9050424. [PMID: 32423149 PMCID: PMC7278598 DOI: 10.3390/antiox9050424] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/12/2020] [Accepted: 05/12/2020] [Indexed: 12/14/2022] Open
Abstract
Antioxidant dietary intervention is considered a potential strategy in delaying age-related dysfunctions. In this study of 56 days, we assessed the antioxidant effects of walnut kernel (WK) and walnut septum extract (WSE) in a D-galactose (D-gal)-induced aging model and in a naturally aged rat model. Young Wistar rats, treated with D-gal (1200 mg/week), and old rats received daily WK or WSE added to the feed. After 8 weeks, blood, liver, and brain samples were collected and hematological, biochemical, oxidative stress biomarkers, histological, and immunohistochemical analyses were performed. Moreover, acetylcholinesterase activity was investigated in brain homogenates. The outcomes demonstrated significant improvement in cellular antioxidant activity and/or decrease of reactive oxygen species, advanced glycation end products, nitric oxide, malondialdehyde, or increase of glutathione after WK or WSE intake in both models. Additionally, WSE showed hypoglycemic effect, and both WK and WSE lowered acetylcholinesterase activity. Both diets could protect neurons against the induced senescence and could reverse the pathological conditions in the physiological aged brain. Thus, dietary supplementation with WK or WSE can maintain the liver and brain health and reduce the risk of age-related diseases, as well as delaying the onset of aging processes.
Collapse
|
35
|
Onaolapo OJ, Adeyemi OI, Amujoyegbe OJ, Fasola EA, Olofinnade AT, Onaolapo AY. High Dietary Fat Modulates Neurobehavioural Effect of Lopinavir/ Ritonavir in Mice. Curr Pharm Biotechnol 2019; 21:158-168. [PMID: 31612827 DOI: 10.2174/1389201020666191011144930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 09/06/2019] [Accepted: 09/24/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Lopinavir/Ritonavir (LR) is a protease inhibitor used human immunodeficiency virus infection management. There have been issues regarding the effects of fat on LR efficacy and the possibility of neurological deficits following prolonged use, there is however a dearth of research examining this. AIMS The effects of LR administered with normal or High-Fat Diet (HFD) on neurobehaviour, neurochemistry and oxidative stress in healthy mice were examined. METHODS Mice were randomly-assigned into eight groups of ten (n=10) animals each. The groups were normal control [Standard Diet, (SD)], HFD control, 3 groups of LR incorporated into SD (100/25, 200/50 and 400/100 mg/kg of feed), and 3 groups of LR with HFD (100/25, 200/50 and 400/100 mg/kg of feed). Mice were fed daily for six weeks, following which open field, elevated-plus maze (EPM), radial-arm maze and Y-maze behaviours were scored. Twenty-four hours after tests, mice were euthanised and brains were homogenised for estimation of oxidative stress, L-glutamate level and acetylcholinesterase activity. RESULTS LR was associated with a reduction in HFD-induced weight gain, suppression of open-field behaviours with SD, and counteraction of HFD-induced changes in working-memory, open-field and anxiety-related behaviours. Also, LR causes increased lipid peroxidation and superoxide dismutase activity; and a decrease in brain glutamate, irrespective of dietary composition. Increased fat catabolism leading to increased oxidative stress could possibly account for the weight changes, while a decrease in brain glutamate could account for the changes in open-field behaviours in mice fed SD. CONCLUSION LR alters neurobehaviour, oxidative stress and brain glutamate in mice; however, only its effects on neurobehaviour are affected by diet.
Collapse
Affiliation(s)
- Olakunle J Onaolapo
- Behavioural Neuroscience and Neuropharmacology Unit, Department of Pharmacology, Ladoke Akintola University of Technology, Osogbo, Osun State, Nigeria
| | - Oluwole I Adeyemi
- Department of Pharmacology, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria
| | - Omolola J Amujoyegbe
- Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Eunice A Fasola
- Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Anthony T Olofinnade
- Department of Pharmacology, Therapeutics and Toxicology, Faculty of Basic Clinical Sciences, College of Medicine, Lagos State University, Lagos state Ikeja, Ikeja, Nigeria
| | - Adejoke Y Onaolapo
- Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria.,Behavioural Neuroscience and Neurobiology Unit, Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| |
Collapse
|
36
|
Lam V, Stephenson A, Nesbit M, Mamsa S, Hackett M, Takechi R, Mamo JCL. Chronic high fat feeding paradoxically attenuates cerebral capillary dysfunction and neurovascular inflammation in Senescence-Accelerated-Murine-Prone Strain 8 mice. Nutr Neurosci 2019; 24:635-643. [PMID: 31510891 DOI: 10.1080/1028415x.2019.1664533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Background: A body of epidemiological, clinical and preclinical studies suggest increased risk for cerebro- and cardio-vascular disease associated with dietary ingestion of long-chain saturated fatty acids (LCSFA). In wild-type rodent models, chronic ingestion of LCSFA diets are associated with increased cerebral capillary permeability, heightened neurovascular inflammation and poorer cognitive performance. However, recent studies suggest that diets enriched in fat may paradoxically attenuate elements of the ageing phenotype via a caloric support axis.Objective: The purpose of this study was to explore the effects of dietary LCSFA on cerebral capillary integrity and neurovascular inflammation in an established model of accelerated ageing, Senescence-Accelerated-Murine-Prone Strain 8 (SAMP8) mice.Methods: From 6 weeks of age, SAMP8 mice and age-matched controls were randomised to either normal chow, or to an LCSFA-enriched diet, for either 12 or 34 weeks. An additional group of SAMP8 mice were provided the LCSFA-enriched diet for 12 weeks followed by the provision of ordinary low-fat chow for 22 weeks. Ex vivo measures of cerebrovascular integrity, neurovascular inflammation and astrocytic activation, were determined via 3-dimensional immunofluorescent confocal microscopy methodologies.Results: LCSFA-fed SAMP8 mice had markedly attenuated cerebral capillary dysfunction concomitant with reduced microglial activation. In SAMP8 mice transiently maintained on an LCSFA diet for 12 weeks, suppression of neurovascular inflammation persisted. Marked hippocampal astrogliosis was evident in LCSFA-fed mice when compared to SAMP8 mice maintained on ordinary chow.Conclusion: The findings from this study support the notion that high-fat, potentially ketogenic diets, may confer neuroprotection in SAMP8 mice through a vascular-support axis.
Collapse
Affiliation(s)
- Virginie Lam
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, WA, Australia.,School of Public Health, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Andrea Stephenson
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, WA, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Michael Nesbit
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, WA, Australia.,School of Public Health, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Somayra Mamsa
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Mark Hackett
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, WA, Australia.,School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Perth, WA, Australia
| | - Ryusuke Takechi
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, WA, Australia.,School of Public Health, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - John C L Mamo
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, WA, Australia.,School of Public Health, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| |
Collapse
|
37
|
Elhaik Goldman S, Goez D, Last D, Naor S, Liraz Zaltsman S, Sharvit-Ginon I, Atrakchi-Baranes D, Shemesh C, Twitto-Greenberg R, Tsach S, Lotan R, Leikin-Frenkel A, Shish A, Mardor Y, Schnaider Beeri M, Cooper I. High-fat diet protects the blood-brain barrier in an Alzheimer's disease mouse model. Aging Cell 2018; 17:e12818. [PMID: 30079520 PMCID: PMC6156545 DOI: 10.1111/acel.12818] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 06/12/2018] [Accepted: 06/24/2018] [Indexed: 12/13/2022] Open
Abstract
Type 2 diabetes (T2D) is associated with increased risk of Alzheimer's disease (AD). There is evidence for impaired blood-brain barrier (BBB) in both diseases, but its role in the interplay between them is not clear. Here, we investigated the effects of high-fat diet (HFD), a model for T2D, on the Tg2576 mouse model of AD, in regard to BBB function. We showed that HFD mice had higher weight, more insulin resistance, and higher serum HDL cholesterol levels, primarily in Tg2576 mice, which also had higher brain lipids content. In terms of behavior, Tg2576 HFD mice were less active and more anxious, but had better learning in the Morris Water Maze compared to Tg2576 on regular diet. HFD had no effect on the level of amyloid beta 1-42 in the cortex of Tg2576 mice, but increased the transcription level of insulin receptor in the hippocampus. Tg2576 mice on regular diet demonstrated more BBB disruption at 8 and 12 months accompanied by larger lateral ventricles volume in contrast to Tg2576 HFD mice, whose BBB leakage and ventricular volume were similar to wild-type (WT) mice. Our results suggest that in AD, HFD may promote better cognitive function through improvements of BBB function and of brain atrophy but not of amyloid beta levels. Lipid metabolism in the CNS and peripheral tissues and brain insulin signaling may underlie this protection.
Collapse
Affiliation(s)
- Shirin Elhaik Goldman
- The Joseph Sagol Neuroscience Center, Sheba Medical Center; Tel Hashomer; Ramat Gan Israel
- Gonda Brain Research Center; Bar Ilan University; Ramat-Gan Israel
| | - David Goez
- The Advanced Technology Center, Sheba Medical Center; Tel-Hashomer; Ramat-Gan Israel
| | - David Last
- The Advanced Technology Center, Sheba Medical Center; Tel-Hashomer; Ramat-Gan Israel
| | - Sharone Naor
- The Joseph Sagol Neuroscience Center, Sheba Medical Center; Tel Hashomer; Ramat Gan Israel
| | - Sigal Liraz Zaltsman
- The Joseph Sagol Neuroscience Center, Sheba Medical Center; Tel Hashomer; Ramat Gan Israel
- Pharmacology Division, Faculty of Medicine, The Institute for Drug Research, School of Pharmacy; Hebrew University of Jerusalem; Jerusalem Israel
| | - Inbal Sharvit-Ginon
- The Joseph Sagol Neuroscience Center, Sheba Medical Center; Tel Hashomer; Ramat Gan Israel
- Department of Psychology; Bar Ilan University; Ramat-Gan Israel
| | - Dana Atrakchi-Baranes
- The Joseph Sagol Neuroscience Center, Sheba Medical Center; Tel Hashomer; Ramat Gan Israel
| | - Chen Shemesh
- The Joseph Sagol Neuroscience Center, Sheba Medical Center; Tel Hashomer; Ramat Gan Israel
| | - Rachel Twitto-Greenberg
- The Joseph Sagol Neuroscience Center, Sheba Medical Center; Tel Hashomer; Ramat Gan Israel
- The Bert W. Strassburger Lipid Center, Sheba Medical Center; Tel-Hashomer; Ramat-Gan Israel
| | - Shoval Tsach
- The Joseph Sagol Neuroscience Center, Sheba Medical Center; Tel Hashomer; Ramat Gan Israel
| | - Roni Lotan
- The Joseph Sagol Neuroscience Center, Sheba Medical Center; Tel Hashomer; Ramat Gan Israel
| | - Alicia Leikin-Frenkel
- The Bert W. Strassburger Lipid Center, Sheba Medical Center; Tel-Hashomer; Ramat-Gan Israel
- Sackler Faculty of Medicine; Tel-Aviv University; Tel-Aviv Israel
| | - Aviv Shish
- The Bert W. Strassburger Lipid Center, Sheba Medical Center; Tel-Hashomer; Ramat-Gan Israel
| | - Yael Mardor
- The Advanced Technology Center, Sheba Medical Center; Tel-Hashomer; Ramat-Gan Israel
- Sackler Faculty of Medicine; Tel-Aviv University; Tel-Aviv Israel
| | - Michal Schnaider Beeri
- The Joseph Sagol Neuroscience Center, Sheba Medical Center; Tel Hashomer; Ramat Gan Israel
- Department of Psychiatry; The Icahn School of Medicine at Mount Sinai; New York New York
- The Interdisciplinary Center; Herzliya Israel
| | - Itzik Cooper
- The Joseph Sagol Neuroscience Center, Sheba Medical Center; Tel Hashomer; Ramat Gan Israel
- The Interdisciplinary Center; Herzliya Israel
| |
Collapse
|