1
|
Asimakidou E, Saipuljumri EN, Lo CH, Zeng J. Role of metabolic dysfunction and inflammation along the liver-brain axis in animal models with obesity-induced neurodegeneration. Neural Regen Res 2025; 20:1069-1076. [PMID: 38989938 PMCID: PMC11438328 DOI: 10.4103/nrr.nrr-d-23-01770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 04/26/2024] [Indexed: 07/12/2024] Open
Abstract
The interaction between metabolic dysfunction and inflammation is central to the development of neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. Obesity-related conditions like type 2 diabetes and non-alcoholic fatty liver disease exacerbate this relationship. Peripheral lipid accumulation, particularly in the liver, initiates a cascade of inflammatory processes that extend to the brain, influencing critical metabolic regulatory regions. Ceramide and palmitate, key lipid components, along with lipid transporters lipocalin-2 and apolipoprotein E, contribute to neuroinflammation by disrupting blood-brain barrier integrity and promoting gliosis. Peripheral insulin resistance further exacerbates brain insulin resistance and neuroinflammation. Preclinical interventions targeting peripheral lipid metabolism and insulin signaling pathways have shown promise in reducing neuroinflammation in animal models. However, translating these findings to clinical practice requires further investigation into human subjects. In conclusion, metabolic dysfunction, peripheral inflammation, and insulin resistance are integral to neuroinflammation and neurodegeneration. Understanding these complex mechanisms holds potential for identifying novel therapeutic targets and improving outcomes for neurodegenerative diseases.
Collapse
Affiliation(s)
- Evridiki Asimakidou
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Eka Norfaishanty Saipuljumri
- School of Applied Science, Republic Polytechnic, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Chih Hung Lo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jialiu Zeng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
2
|
Zhou AJ, Xiong ZE, Wang L, Chen XX, Wang ZP, Zhang YD, Chen WW, Cai XL, Xu YL, Rong S, Wang T. Long-Term Administration of Nicotinamide Mononucleotide Mitigates High-Fat-Diet-Induced Physiological Decline in Aging Mice. J Nutr 2024:S0022-3166(24)01094-0. [PMID: 39424071 DOI: 10.1016/j.tjnut.2024.10.017] [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: 06/16/2024] [Revised: 09/24/2024] [Accepted: 10/02/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Nicotinamide adenine dinucleotide (NAD+) levels decline with age, and boosting it can improve multi-organ functions and lifespan. OBJECTIVES Nicotinamide mononucleotide (NMN) is a natural NAD+ precursor with the ability to enhance NAD+ biosynthesis. Numerous studies have shown that a high-fat diet (HFD) can accelerate the process of aging and many diseases. We hypothesized that long-term administration of NMN could exert protective effects on adipose, muscle, and kidney tissues in mice on an HFD act by affecting the autophagic pathway. METHODS Mice at 14 mo of age were fed an HFD, and NMN was added to their drinking water at a dose of 400 mg/kg for 7 mo. The locomotor ability of the mice was assessed by behavioral experiments such as grip test, wire hang test, rotarod, and beam-walking test. At the end of the behavioral experiments, the pathological changes of each peripheral organ and the expression of autophagy-related proteins, as well as the markers of the senescence and inflammaging were analyzed by pathological staining, immunohistochemical staining, and western blotting, respectively. RESULTS We found that NMN supplementation increased NAD+ levels and ultimately attenuated age- and diet-related physiological decline in mice. NMN inhibited HFD-induced obesity, promoted physical activity, improved glucose and lipid metabolism, improved skeletal muscle function and renal damage, as well as mitigated the senescence and inflammaging as demonstrated by p16, interleukin 1β, and tumor necrosis factor α levels. In addition, the present study further emphasizes the potential mechanisms underlying the bidirectional relationship between NAD+ and autophagy. We detected changes in autophagy levels in various tissue organs, and NMN may play a protective role by inhibiting excessive autophagy induced by HFD. CONCLUSIONS Our findings demonstrated that NMN administration attenuated HFD-induced metabolic disorders and physiological decline in aging mice.
Collapse
Affiliation(s)
- Ao-Jia Zhou
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China; Department of Pharmacy, Institute of Pharmaceutical Process, College of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Zhang-E Xiong
- Department of Gastroenterology, Wuhan Third Hospital, Wuhan, China
| | - Li Wang
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China; Department of Pharmacy, Institute of Pharmaceutical Process, College of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Xiao-Xuan Chen
- Department of Pharmacy, Institute of Pharmaceutical Process, College of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Zi-Ping Wang
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China
| | - Yi-Dan Zhang
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China
| | - Wen-Wen Chen
- Department of Pharmacy, Institute of Pharmaceutical Process, College of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Xiao-Li Cai
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China
| | - Yang-Liu Xu
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China
| | - Shuang Rong
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China; Department of Food and Nutrition Health, School of Public Health, Wuhan University, Wuhan, China.
| | - Ting Wang
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China; Department of Pharmacy, Institute of Pharmaceutical Process, College of Medicine, Wuhan University of Science and Technology, Wuhan, China; Wuhan Asia General Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, China.
| |
Collapse
|
3
|
Mackey-Alfonso SE, Butler MJ, Taylor AM, Williams-Medina AR, Muscat SM, Fu H, Barrientos RM. Short-term high fat diet impairs memory, exacerbates the neuroimmune response, and evokes synaptic degradation via a complement-dependent mechanism in a mouse model of Alzheimer's disease. Brain Behav Immun 2024; 121:56-69. [PMID: 39043341 DOI: 10.1016/j.bbi.2024.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 07/25/2024] Open
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disease characterized by profound memory impairments, synaptic loss, neuroinflammation, and hallmark pathological markers. High-fat diet (HFD) consumption increases the risk of developing AD even after controlling for metabolic syndrome, pointing to a role of the diet itself in increasing risk. In AD, the complement system, an arm of the immune system which normally tags redundant or damaged synapses for pruning, becomes pathologically overactivated leading to tagging of healthy synapses. While the unhealthy diet to AD link is strong, the underlying mechanisms are not well understood in part due to confounding variables associated with long-term HFD which can independently influence the brain. Therefore, we experimented with a short-term diet regimen to isolate the diet's impact on brain function without causing obesity. This project investigated the effect of short-term HFD on 1) memory, 2) neuroinflammation including complement, 3) AD pathology markers, 4) synaptic markers, and 5) in vitro microglial synaptic phagocytosis in the 3xTg-AD mouse model. Following the consumption of either standard chow or HFD, 3xTg-AD and non-Tg mice were tested for memory impairments. In a separate cohort of mice, levels of hippocampal inflammatory markers, complement proteins, AD pathology markers, and synaptic markers were measured. For the last set of experiments, BV2 microglial phagocytosis of synapses was evaluated. Synaptoneurosomes isolated from the hippocampus of 3xTg-AD mice fed chow or HFD were incubated with equal numbers of BV2 microglia. The number of BV2 microglia that phagocytosed synaptoneurosomes was tracked over time with a live-cell imaging assay. Finally, we incubated BV2 microglia with a complement receptor inhibitor (NIF) and repeated the assay. Behavioral analysis showed 3xTg-AD mice had significantly impaired long-term contextual and cued fear memory compared to non-Tg mice that was further impaired by HFD. HFD significantly increased inflammatory markers and complement expression while decreasing synaptic marker expression only in 3xTg-AD mice, without altering AD pathology markers. Synaptoneurosomes from HFD-fed 3xTg-AD mice were phagocytosed at a significantly higher rate than those from chow-fed mice, suggesting the synapses were altered by HFD. The complement receptor inhibitor blocked this effect in a dose-dependent manner, demonstrating the HFD-mediated increase in phagocytosis was complement dependent. This study indicates HFD consumption increases neuroinflammation and over-activates the complement cascade in 3xTg-AD mice, resulting in poorer memory. The in vitro data point to complement as a potential mechanistic culprit and therapeutic target underlying HFD's influence in increasing cognitive vulnerability to AD.
Collapse
Affiliation(s)
- Sabrina E Mackey-Alfonso
- Medical Scientist Training Program, The Ohio State University, Columbus, OH, USA; Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA; Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Michael J Butler
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA; Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, USA
| | - Ashton M Taylor
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | | | - Stephanie M Muscat
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Hongjun Fu
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA; Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
4
|
Zhao N, Huang Y, Cheng X, Xie L, Xiao W, Shi B, Li J. A critical size volumetric muscle loss model in mouse masseter with impaired mastication on nutrition. Cell Prolif 2024; 57:e13610. [PMID: 38356342 DOI: 10.1111/cpr.13610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/13/2024] [Accepted: 01/30/2024] [Indexed: 02/16/2024] Open
Abstract
Orofacial muscle defect due to congenital anomalies, tumour ablation or traumatic accident that exceeds endogenous regeneration capacity may lead to sustained deficits in masticatory function and nutrition intake. Functional recovery has always been the goal of muscle tissue repair, but currently, there is no suitable model for quantitative analyses of either functional consequences or treatment efficacy of orofacial muscle defect. This study proposed a critical size volumetric muscle loss (VML) model in mouse masseter with impaired mastication on nutrition. Full-thickness VML defects in diameter of 1.0, 1.5, 2.0 and 3.0 mm were generated in the centre of the mouse masseter using a biopsy punch to determine the critical size for functional impairment. In the VML region, myogenesis was dampened but fibrogenesis was activated, as long with a reduction in the density of the neuromuscular junction and an increase in vascular density. Accordingly, persistent fibrosis was observed in the centre region of VML in all diameters. The 2.0 mm diameter was the critical threshold to masticatory function impairment after VML in the masseter. VML of 3.0 mm diameter led to a significant impact on nutrition intake and body weight gain. Autologous muscle graft effectively relieved the fibrosis and functional deficit after VML injury in the masseter. This model serves as a reliable tool in studying functional recovery strategies for orofacial muscle defects.
Collapse
Affiliation(s)
- Ning Zhao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yixuan Huang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xu Cheng
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| | - Li Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
| | - Wenlin Xiao
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bing Shi
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jingtao Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Jang YJ, Choi MG, Yoo BJ, Lee KJ, Jung WB, Kim SG, Park SA. Interaction Between a High-Fat Diet and Tau Pathology in Mice: Implications for Alzheimer's Disease. J Alzheimers Dis 2024; 97:485-506. [PMID: 38108353 DOI: 10.3233/jad-230927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
BACKGROUND Obesity is a modifiable risk factor for Alzheimer's disease (AD). However, its relation with tau pathology (i.e., aberrant tau protein behavior in tauopathies such as AD) has been inconclusive. OBJECTIVE This study investigated the interaction between a high-fat diet (HFD) and tau pathology in adult male mice. METHODS Transgenic mice overexpressing human P301S Tau (those with the pathology) and wild-type (WT) littermates were subjected to behavioral tests, functional magnetic resonance imaging (fMRI), diffusion tensor imaging (DTI), and western blotting analysis to investigate the effects of prolonged HFD versus regular diet during adulthood. RESULTS HFD increased body weight in both WT and P301S mice but had minimal effect on blood glucose levels. The brain response to HFD was tau genotype-specific. WT mice exhibited decreased recognition memory and enhanced network connectivity in fMRI, while P301S mice exhibited white matter tract disorganization in DTI as the sole significant finding. The reduction of insulin receptor β, insulin downstream signaling, neuronal nuclear protein, CD68-positive phagocytic activity, and myelin basic protein level were confined to the cortex of WT mice. In contrast to P301S mice, WT mice showed significant changes in the tau protein and its phosphorylation levels along with increased soluble neurofilament light levels in the hippocampus. CONCLUSIONS HFD-induced brain dysfunction and pathological changes were blunted in mice with the pathology and more profound in healthy mice. Our findings highlight the need to consider this interaction between obesity and tau pathology when tailoring treatment strategies for AD and other tauopathies.
Collapse
Affiliation(s)
- Yu Jung Jang
- Lab for Neurodegenerative Dementia, Department of Anatomy, Ajou University School of Medicine, Suwon, Republic of Korea
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Republic of Korea
| | - Min Gyu Choi
- Lab for Neurodegenerative Dementia, Department of Anatomy, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Byung Jae Yoo
- Lab for Neurodegenerative Dementia, Department of Anatomy, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Kyeong Jae Lee
- Lab for Neurodegenerative Dementia, Department of Anatomy, Ajou University School of Medicine, Suwon, Republic of Korea
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Republic of Korea
| | - Won Beom Jung
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon, Republic of Korea
| | - Seong-Gi Kim
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon, Republic of Korea
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sun Ah Park
- Lab for Neurodegenerative Dementia, Department of Anatomy, Ajou University School of Medicine, Suwon, Republic of Korea
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Republic of Korea
- Department of Neurology, Ajou University School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
6
|
Yang K, Hou R, Zhao J, Wang X, Wei J, Pan X, Zhu X. Lifestyle effects on aging and CVD: A spotlight on the nutrient-sensing network. Ageing Res Rev 2023; 92:102121. [PMID: 37944707 DOI: 10.1016/j.arr.2023.102121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/12/2023] [Accepted: 11/04/2023] [Indexed: 11/12/2023]
Abstract
Aging is widespread worldwide and a significant risk factor for cardiovascular disease (CVD). Mechanisms underlying aging have attracted considerable attention in recent years. Remarkably, aging and CVD overlap in numerous ways, with deregulated nutrient sensing as a common mechanism and lifestyle as a communal modifier. Interestingly, lifestyle triggers or suppresses multiple nutrient-related signaling pathways. In this review, we first present the composition of the nutrient-sensing network (NSN) and its metabolic impact on aging and CVD. Secondly, we review how risk factors closely associated with CVD, including adverse life states such as sedentary behavior, sleep disorders, high-fat diet, and psychosocial stress, contribute to aging and CVD, with a focus on the bridging role of the NSN. Finally, we focus on the positive effects of beneficial dietary interventions, specifically dietary restriction and the Mediterranean diet, on the regulation of nutrient metabolism and the delayed effects of aging and CVD that depend on the balance of the NSN. In summary, we expound on the interaction between lifestyle, NSN, aging, and CVD.
Collapse
Affiliation(s)
- Kaiying Yang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Rongyao Hou
- Department of Neurology, The Affiliated Hiser Hospital of Qingdao University, Qingdao 266000, China
| | - Jie Zhao
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xia Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jin Wei
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
7
|
Lippi SLP, Barkey RE, Rodriguez MN. High-fat diet negatively affects brain markers, cognitive behaviors, and noncognitive behaviors in the rTg4510 tau mouse model. Physiol Behav 2023; 271:114316. [PMID: 37543107 DOI: 10.1016/j.physbeh.2023.114316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/11/2023] [Accepted: 08/01/2023] [Indexed: 08/07/2023]
Abstract
Alzheimer's disease (AD) drastically impacts cognitive and noncognitive behaviors in both humans and animal models. Two hallmark proteins in AD, amyloid-β plaques and tau neurofibrillary tangles, accumulate in regions of the brain critical for learning and memory, including the hippocampus. Poor dietary choices have been shown to exacerbate cognitive deficits seen in AD. In this study, we assessed the effects of a high-fat diet (HFD - 60 kcal% fat) on cognitive & noncognitive behaviors as well as on brain markers in the rTg4510 tau mouse model. While all mice learned the Morris Water Maze (MWM) task, it was noted that on the last day of acquisition female tau mice had a significantly higher latency to find the platform than male tau mice (p < 0.01). Mice given the HFD spent significantly less time in the target quadrant than those given a control diet (CD) (p < 0.05). Tau mice showed impaired burrowing (p < 0.05) and nesting behaviors (p < 0.001) compared to WT mice and HFD administration worsened burrowing in tau mice. Tau mice exhibited greater levels of glial fibrillary acidic protein (GFAP) (p < 0.05) and significantly less hippocampal cell density than WT mice (p < 0.001). We observed trends of HFD mice having greater levels of GFAP and greater average tangle size than CD mice. These results highlight the importance of dietary choices, especially in older populations more susceptible to AD and its effects.
Collapse
Affiliation(s)
- Stephen L P Lippi
- University of Texas at San Antonio, Dept. Psychology, San Antonio, TX 78249, United States.
| | - Rachel E Barkey
- Pennsylvania State University College of Medicine, Dept. Neural and Behavioral Sciences, 700 HMC Crescent Road, Hershey, PA 17033, United States
| | - Mya N Rodriguez
- MD Anderson UTHealth Houston Graduate School of Biomedical Sciences, 6767 Bertner Ave, Houston, TX 77030, United States
| |
Collapse
|
8
|
Wang F, Wan J, Liao Y, Liu S, Wei Y, Ouyang Z. Dendrobium species regulate energy homeostasis in neurodegenerative diseases: a review. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2023.03.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
9
|
Mengr A, Strnadová V, Strnad Š, Vrkoslav V, Pelantová H, Kuzma M, Comptdaer T, Železná B, Kuneš J, Galas MC, Pačesová A, Maletínská L. Feeding High-Fat Diet Accelerates Development of Peripheral and Central Insulin Resistance and Inflammation and Worsens AD-like Pathology in APP/PS1 Mice. Nutrients 2023; 15:3690. [PMID: 37686722 PMCID: PMC10490051 DOI: 10.3390/nu15173690] [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: 07/13/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 09/10/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive brain disorder characterized by extracellular amyloid-β (Aβ) plaques, intracellular neurofibrillary tangles formed by hyperphosphorylated Tau protein and neuroinflammation. Previous research has shown that obesity and type 2 diabetes mellitus, underlined by insulin resistance (IR), are risk factors for neurodegenerative disorders. In this study, obesity-induced peripheral and central IR and inflammation were studied in relation to AD-like pathology in the brains and periphery of APP/PS1 mice, a model of Aβ pathology, fed a high-fat diet (HFD). APP/PS1 mice and their wild-type controls fed either a standard diet or HFD were characterized at the ages of 3, 6 and 10 months by metabolic parameters related to obesity via mass spectroscopy, nuclear magnetic resonance, immunoblotting and immunohistochemistry to quantify how obesity affected AD pathology. The HFD induced substantial peripheral IR leading to central IR. APP/PS1-fed HFD mice had more pronounced IR, glucose intolerance and liver steatosis than their WT controls. The HFD worsened Aβ pathology in the hippocampi of APP/PS1 mice and significantly supported both peripheral and central inflammation. This study reveals a deleterious effect of obesity-related mild peripheral inflammation and prediabetes on the development of Aβ and Tau pathology and neuroinflammation in APP/PS1 mice.
Collapse
Affiliation(s)
- Anna Mengr
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, Prague 6, 166 10 Prague, Czech Republic; (A.M.); (V.S.); (Š.S.); (V.V.); (B.Ž.); (J.K.)
| | - Veronika Strnadová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, Prague 6, 166 10 Prague, Czech Republic; (A.M.); (V.S.); (Š.S.); (V.V.); (B.Ž.); (J.K.)
| | - Štěpán Strnad
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, Prague 6, 166 10 Prague, Czech Republic; (A.M.); (V.S.); (Š.S.); (V.V.); (B.Ž.); (J.K.)
| | - Vladimír Vrkoslav
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, Prague 6, 166 10 Prague, Czech Republic; (A.M.); (V.S.); (Š.S.); (V.V.); (B.Ž.); (J.K.)
| | - Helena Pelantová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 4, 142 20 Prague, Czech Republic; (H.P.); (M.K.)
| | - Marek Kuzma
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 4, 142 20 Prague, Czech Republic; (H.P.); (M.K.)
| | - Thomas Comptdaer
- University of Lille, Inserm, CHU Lille, CNRS, LilNCog-Lille Neuroscience & Cognition, F-59000 Lille, France; (T.C.); (M.-C.G.)
| | - Blanka Železná
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, Prague 6, 166 10 Prague, Czech Republic; (A.M.); (V.S.); (Š.S.); (V.V.); (B.Ž.); (J.K.)
| | - Jaroslav Kuneš
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, Prague 6, 166 10 Prague, Czech Republic; (A.M.); (V.S.); (Š.S.); (V.V.); (B.Ž.); (J.K.)
- Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 4, 142 20 Prague, Czech Republic
| | - Marie-Christine Galas
- University of Lille, Inserm, CHU Lille, CNRS, LilNCog-Lille Neuroscience & Cognition, F-59000 Lille, France; (T.C.); (M.-C.G.)
| | - Andrea Pačesová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, Prague 6, 166 10 Prague, Czech Republic; (A.M.); (V.S.); (Š.S.); (V.V.); (B.Ž.); (J.K.)
| | - Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, Prague 6, 166 10 Prague, Czech Republic; (A.M.); (V.S.); (Š.S.); (V.V.); (B.Ž.); (J.K.)
| |
Collapse
|
10
|
Gong Y, Luo H, Li Z, Feng Y, Liu Z, Chang J. Metabolic Profile of Alzheimer's Disease: Is 10-Hydroxy-2-decenoic Acid a Pertinent Metabolic Adjuster? Metabolites 2023; 13:954. [PMID: 37623897 PMCID: PMC10456792 DOI: 10.3390/metabo13080954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/12/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023] Open
Abstract
Alzheimer's disease (AD) represents a significant public health concern in modern society. Metabolic syndrome (MetS), which includes diabetes mellitus (DM) and obesity, represents a modifiable risk factor for AD. MetS and AD are interconnected through various mechanisms, such as mitochondrial dysfunction, oxidative stress, insulin resistance (IR), vascular impairment, inflammation, and endoplasmic reticulum (ER) stress. Therefore, it is necessary to seek a multi-targeted and safer approach to intervention. Thus, 10-hydroxy-2-decenoic acid (10-HDA), a unique hydroxy fatty acid in royal jelly, has shown promising anti-neuroinflammatory, blood-brain barrier (BBB)-preserving, and neurogenesis-promoting properties. In this paper, we provide a summary of the relationship between MetS and AD, together with an introduction to 10-HDA as a potential intervention nutrient. In addition, molecular docking is performed to explore the metabolic tuning properties of 10-HDA with associated macromolecules such as GLP-1R, PPARs, GSK-3, and TREM2. In conclusion, there is a close relationship between AD and MetS, and 10-HDA shows potential as a beneficial nutritional intervention for both AD and MetS.
Collapse
Affiliation(s)
| | | | | | | | | | - Jie Chang
- Department of Occupational and Environmental Health, School of Public Health, Soochow University, 199 Ren’ai Road, Suzhou 215123, China; (Y.G.)
| |
Collapse
|
11
|
Yang S, Wang L, Zeng Y, Wang Y, Pei T, Xie Z, Xiong Q, Wei H, Li W, Li J, Su Q, Wei D, Cheng W. Salidroside alleviates cognitive impairment by inhibiting ferroptosis via activation of the Nrf2/GPX4 axis in SAMP8 mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154762. [PMID: 36965372 DOI: 10.1016/j.phymed.2023.154762] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 03/03/2023] [Accepted: 03/11/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurogenerative disease and remains no effective method for stopping its progress. Ferroptosis and adaptive immunity have been proven to contribute to AD pathogenesis. Salidroside exhibits neuroprotective and immunomodulatory effects. However, the underlying mechanisms linking salidroside, ferroptosis, and adaptive immunity in AD remain uncertain. PURPOSE The objective of this study is to explore the neuroprotective effects and the potential molecular mechanisms of salidroside against neuronal ferroptosis and CD8+ T cell infiltration in senescence-accelerated mouse prone 8 (SAMP8) mice. STUDY DESIGN AND METHODS SAMP8 mice were employed as an AD model and were treated with salidroside for 12 weeks. Behavioral tests, immunohistochemistry, HE and Nissl staining, immunofluorescence, transmission electron microscopy, quantitative proteomics, bioinformatic analysis, flow cytometry, iron staining, western blotting, and molecular docking were performed. RESULTS Treatment with salidroside dose-dependently attenuated cognitive impairment, reduced the accumulation of Aβ plaques and restored neuronal damage. Salidroside also suppressed the infiltration of CD8+T cells, oxidative stress, and inflammatory cytokines, and improved mitochondrial metabolism, iron metabolism, lipid metabolism, and redox in the SAMP8 mice brain. The administration of salidroside decreased iron deposition, reduced TFR1, and ACSL4 protein expression, upregulated SLC7A11, and GPX4 protein expression, and promoted the Nrf2/GPX4 axis activation. CONCLUSION In conclusion, neuronal ferroptosis and CD8+T cells are involved in the process of cognitive impairment in SAMP8 mice. Salidroside alleviates cognitive impairment and inhibits neuronal ferroptosis. The underlying mechanisms may involve the Nrf2/GPX4 axis activation and reduction in CD8+T cells infiltration. This study provides some evidence for the roles of salidroside in adaptive immunity and neuronal ferroptosis in SAMP8 mice.
Collapse
Affiliation(s)
- Sixia Yang
- Department of Pharmacy, Zhu Jiang Hospital, Southern Medical University, Guangzhou 510260, China; School of Traditional Chinese Medicine, Southern Medical University, No.1838, North Guangzhou Avenue, Baiyun District, Guangzhou 510515, China
| | - Linshuang Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, No. 16, Nanxiao Street, Dongzhimen Nei, Dongcheng District, Beijing 100700, China
| | - Yi Zeng
- School of Traditional Chinese Medicine, Southern Medical University, No.1838, North Guangzhou Avenue, Baiyun District, Guangzhou 510515, China
| | - Yong Wang
- Department of Pharmacy, Zhu Jiang Hospital, Southern Medical University, Guangzhou 510260, China
| | - Tingting Pei
- School of Traditional Chinese Medicine, Southern Medical University, No.1838, North Guangzhou Avenue, Baiyun District, Guangzhou 510515, China
| | - Zeping Xie
- School of Traditional Chinese Medicine, Southern Medical University, No.1838, North Guangzhou Avenue, Baiyun District, Guangzhou 510515, China
| | - Qiaowu Xiong
- School of Traditional Chinese Medicine, Southern Medical University, No.1838, North Guangzhou Avenue, Baiyun District, Guangzhou 510515, China
| | - Hui Wei
- School of Traditional Chinese Medicine, Southern Medical University, No.1838, North Guangzhou Avenue, Baiyun District, Guangzhou 510515, China
| | - Wenxu Li
- School of Traditional Chinese Medicine, Southern Medical University, No.1838, North Guangzhou Avenue, Baiyun District, Guangzhou 510515, China
| | - Jiaqi Li
- School of Traditional Chinese Medicine, Southern Medical University, No.1838, North Guangzhou Avenue, Baiyun District, Guangzhou 510515, China
| | - Qian Su
- School of Traditional Chinese Medicine, Southern Medical University, No.1838, North Guangzhou Avenue, Baiyun District, Guangzhou 510515, China
| | - Dongfeng Wei
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, No. 16, Nanxiao Street, Dongzhimen Nei, Dongcheng District, Beijing 100700, China.
| | - Weidong Cheng
- Department of Pharmacy, Zhu Jiang Hospital, Southern Medical University, Guangzhou 510260, China; School of Traditional Chinese Medicine, Southern Medical University, No.1838, North Guangzhou Avenue, Baiyun District, Guangzhou 510515, China.
| |
Collapse
|
12
|
Na Y, Ke L, Jie Z, Jinping W, Tao M, Jie Z, Liu Y, Yueqin Z. Amelioration of Cholesterol Rich diet-induced Impaired Cognition in AD Transgenic Mice by an LXR Agonist TO901317 Is Associated with the Activation of the LXR-β-RXR-α-ABCA1 Transmembrane Transport System and Improving the Composition of Lipid Raft. Exp Aging Res 2023; 49:214-225. [PMID: 35792710 DOI: 10.1080/0361073x.2022.2095605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 06/26/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND It has been reported that LXR agonist can inhibit Aβ generation and alleviate Aβ-induced various adverse reactions in vivo and in vitro experiments, but the mechanisms have not been clarified. The study aimed to observe the effect of LXR agonist TO901317 on the cognitive function of AD transgenic mice fed with cholesterol-rich diet (CRD), and to explore the possible mechanism. Methods: 32 male 6-month-old double transgenic AD mice were enrolled and randomly divided into 4 groups: control (normal diet) group, CRD treatment group, TO901317 treatment group and GSK2033 treatment group. After 3 month, Morris water maze was for the changes of spatial exploration and memory ability; ELISA was for detecting the production of Aβ42 in the brain; the concentration of total cholesterol (TC), low density lipoprotein (LDL) and high density lipoprotein (HDL) in serum were detected by cholesterol enzyme colorimetry; Finally, the expression of LXR-β, RXR-α, ABCA1, caveolin-1, BACE1 and APP at protein level in the brains was measured by Western blotting. RESULTS Compared with the control group, the learning, memory ability and spatial exploration ability of the mice were more significantly serious in the CRD group (P<0.05); The contents of TC and LDL in the serum and the production of Aβ42 in the brains were significantly increased (P<0.05), but HDL was remarkably decreased (P<0.05); The protein levels of LXR-β, RXR-α and ABCA1 were also significantly decreased (P<0.05); The expression of caveolin-1, APP and BACE1 were evidently increased (P<0.05). However, after treatment with TO901317, the impaired learning and memory and spatial exploration ability of the mice were significantly improved (P<0.05); The contents of TC and LDL in serum and the production of Aβ42 in the brains were significantly decreased (P<0.05), but HLD was increased (P<0.05); The protein levels of LXR-β, RXR-α, ABCA1were all significantly increased (P<0.05), while, the expression of caveolin-1, APP and BACE1 were all significantly decreased (P<0.05). All the changes were reversed by GSK2033 (P<0.05). CONCLUSIONS TO901317 attenuated the more serious impairment of spatial exploration, learning and memory in transgenic AD mice induced by CRD, and the mechanism may be that TO901317 could activate the LXR-β/RXR-α/ABCA1 transmembrane transport system, promote the cholesterol efflux, and decreased caveolin-1, APP and BACE1, further reduce Aβ42 in the brains.
Collapse
Affiliation(s)
- Yang Na
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Lin Ke
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Zhang Jie
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Wang Jinping
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Meng Tao
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Zhu Jie
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Yang Liu
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Zhou Yueqin
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| |
Collapse
|
13
|
Cui SS, Jiang QW, Chen SD. Sex difference in biological change and mechanism of Alzheimer’s disease: from macro- to micro-landscape. Ageing Res Rev 2023; 87:101918. [PMID: 36967089 DOI: 10.1016/j.arr.2023.101918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 02/16/2023] [Accepted: 03/23/2023] [Indexed: 04/05/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia and numerous studies reported a higher prevalence and incidence of AD among women. Although women have longer lifetime, longevity does not wholly explain the higher frequency and lifetime risk in women. It is important to understand sex differences in AD pathophysiology and pathogenesis, which could provide foundation for future clinical AD research. Here, we reviewed the most recent and relevant literature on sex differences in biological change of AD from macroscopical neuroimaging to microscopical pathologic change (neuronal degeneration, synaptic dysfunction, amyloid-beta and tau accumulation). We also discussed sex differences in cellular mechanisms related to AD (neuroinflammation, mitochondria dysfunction, oxygen stress, apoptosis, autophagy, blood-brain-barrier dysfunction, gut microbiome alteration, bulk and single cell/nucleus omics) and possible causes underlying these differences including sex-chromosome, sex hormone and hypothalamic-pituitary- adrenal (HPA) axis effects.
Collapse
Affiliation(s)
- Shi-Shuang Cui
- Department of Neurology & Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Geriatrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qian-Wen Jiang
- Department of Neurology & Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Geriatrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Sheng-Di Chen
- Department of Neurology & Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
14
|
Kelty TJ, Dashek RJ, Arnold WD, Rector RS. Emerging Links between Nonalcoholic Fatty Liver Disease and Neurodegeneration. Semin Liver Dis 2023; 43:77-88. [PMID: 36764305 DOI: 10.1055/s-0043-1762585] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
The association between liver and brain health has gained attention as biomarkers of liver function have been revealed to predict neurodegeneration. The liver is a central regulator in metabolic homeostasis. However, in nonalcoholic fatty liver disease (NAFLD), homeostasis is disrupted which can result in extrahepatic organ pathologies. Emerging literature provides insight into the mechanisms behind the liver-brain health axis. These include the increased production of liver-derived factors that promote insulin resistance and loss of neuroprotective factors under conditions of NAFLD that increase insulin resistance in the central nervous system. In addition, elevated proinflammatory cytokines linked to NAFLD negatively impact the blood-brain barrier and increase neuroinflammation. Furthermore, exacerbated dyslipidemia associated with NAFLD and hepatic dysfunction can promote altered brain bioenergetics and oxidative stress. In this review, we summarize the current knowledge of the crosstalk between liver and brain as it relates to the pathophysiology between NAFLD and neurodegeneration, with an emphasis on Alzheimer's disease. We also highlight knowledge gaps and future areas for investigation to strengthen the potential link between NAFLD and neurodegeneration.
Collapse
Affiliation(s)
- Taylor J Kelty
- Department of Biomedical Sciences, University of Missouri - Columbia, Columbia, Missouri
- Department of Nutrition and Exercise Physiology, University of Missouri - Columbia, Columbia, Missouri
- NextGen Precision Health, University of Missouri - Columbia, Columbia, Missouri
| | - Ryan J Dashek
- Department of Biomedical Sciences, University of Missouri - Columbia, Columbia, Missouri
- NextGen Precision Health, University of Missouri - Columbia, Columbia, Missouri
- Comparative Medicine Program, University of Missouri - Columbia, Columbia, Missouri
| | - W David Arnold
- NextGen Precision Health, University of Missouri - Columbia, Columbia, Missouri
- Physical Medicine and Rehabilitation, University of Missouri - Columbia, Columbia, Missouri
| | - R Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri - Columbia, Columbia, Missouri
- NextGen Precision Health, University of Missouri - Columbia, Columbia, Missouri
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri - Columbia, Columbia, Missouri
| |
Collapse
|
15
|
Shirokova O, Zaborskaya O, Pchelin P, Kozliaeva E, Pershin V, Mukhina I. Genetic and Epigenetic Sexual Dimorphism of Brain Cells during Aging. Brain Sci 2023; 13:brainsci13020195. [PMID: 36831738 PMCID: PMC9954625 DOI: 10.3390/brainsci13020195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/27/2023] Open
Abstract
In recent years, much of the attention paid to theoretical and applied biomedicine, as well as neurobiology, has been drawn to various aspects of sexual dimorphism due to the differences that male and female brain cells demonstrate during aging: (a) a dimorphic pattern of response to therapy for neurodegenerative disorders, (b) different age of onset and different degrees of the prevalence of such disorders, and (c) differences in their symptomatic manifestations in men and women. The purpose of this review is to outline the genetic and epigenetic differences in brain cells during aging in males and females. As a result, we hereby show that the presence of brain aging patterns in males and females is due to a complex of factors associated with the effects of sex chromosomes, which subsequently entails a change in signal cascades in somatic cells.
Collapse
Affiliation(s)
- Olesya Shirokova
- Institute of Fundamental Medicine, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603950, Russia
- Correspondence:
| | - Olga Zaborskaya
- Institute of Fundamental Medicine, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603950, Russia
| | - Pavel Pchelin
- Institute of Fundamental Medicine, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603950, Russia
- Institute of Biology and Biomedicine, Lobachevsky State University, 23 Gagarin Avenue, Nizhny Novgorod 603002, Russia
| | - Elizaveta Kozliaeva
- Institute of Fundamental Medicine, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603950, Russia
| | - Vladimir Pershin
- Institute of Fundamental Medicine, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603950, Russia
- Institute of Biology and Biomedicine, Lobachevsky State University, 23 Gagarin Avenue, Nizhny Novgorod 603002, Russia
| | - Irina Mukhina
- Institute of Fundamental Medicine, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603950, Russia
- Institute of Biology and Biomedicine, Lobachevsky State University, 23 Gagarin Avenue, Nizhny Novgorod 603002, Russia
| |
Collapse
|
16
|
Marcos JL, Olivares-Barraza R, Ceballo K, Wastavino M, Ortiz V, Riquelme J, Martínez-Pinto J, Muñoz P, Cruz G, Sotomayor-Zárate R. Obesogenic Diet-Induced Neuroinflammation: A Pathological Link between Hedonic and Homeostatic Control of Food Intake. Int J Mol Sci 2023; 24:ijms24021468. [PMID: 36674982 PMCID: PMC9866213 DOI: 10.3390/ijms24021468] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 01/13/2023] Open
Abstract
Obesity-induced neuroinflammation is a chronic aseptic central nervous system inflammation that presents systemic characteristics associated with increased pro-inflammatory cytokines such as interleukin 1 beta (IL-1β) and interleukin 18 (IL-18) and the presence of microglia and reactive astrogliosis as well as the activation of the NLRP3 inflammasome. The obesity pandemic is associated with lifestyle changes, including an excessive intake of obesogenic foods and decreased physical activity. Brain areas such as the lateral hypothalamus (LH), lateral septum (LS), ventral tegmental area (VTA), and nucleus accumbens (NAcc) have been implicated in the homeostatic and hedonic control of feeding in experimental models of diet-induced obesity. In this context, a chronic lipid intake triggers neuroinflammation in several brain regions such as the hypothalamus, hippocampus, and amygdala. This review aims to present the background defining the significant impact of neuroinflammation and how this, when induced by an obesogenic diet, can affect feeding control, triggering metabolic and neurological alterations.
Collapse
Affiliation(s)
- José Luis Marcos
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Escuela de Ciencias Agrícolas y Veterinarias, Universidad Viña del Mar, Viña del Mar 2572007, Chile
- Programa de Doctorado en Ciencias e Ingeniería para la Salud, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Rossy Olivares-Barraza
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Programa de Doctorado en Ciencias Mención Neurociencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Karina Ceballo
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Programa de Doctorado en Ciencias Mención Neurociencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Melisa Wastavino
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Víctor Ortiz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Julio Riquelme
- Escuela de Medicina y Centro de Neurología Traslacional (CENTRAS), Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2540064, Chile
| | - Jonathan Martínez-Pinto
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Pablo Muñoz
- Escuela de Medicina y Centro de Neurología Traslacional (CENTRAS), Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2540064, Chile
| | - Gonzalo Cruz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Ramón Sotomayor-Zárate
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Correspondence: ; Tel.: +56-32-2508050
| |
Collapse
|
17
|
de la Monte SM. Malignant Brain Aging: The Formidable Link Between Dysregulated Signaling Through Mechanistic Target of Rapamycin Pathways and Alzheimer's Disease (Type 3 Diabetes). J Alzheimers Dis 2023; 95:1301-1337. [PMID: 37718817 PMCID: PMC10896181 DOI: 10.3233/jad-230555] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Malignant brain aging corresponds to accelerated age-related declines in brain functions eventually derailing the self-sustaining forces that govern independent vitality. Malignant brain aging establishes the path toward dementing neurodegeneration, including Alzheimer's disease (AD). The full spectrum of AD includes progressive dysfunction of neurons, oligodendrocytes, astrocytes, microglia, and the microvascular systems, and is mechanistically driven by insulin and insulin-like growth factor (IGF) deficiencies and resistances with accompanying deficits in energy balance, increased cellular stress, inflammation, and impaired perfusion, mimicking the core features of diabetes mellitus. The underlying pathophysiological derangements result in mitochondrial dysfunction, abnormal protein aggregation, increased oxidative and endoplasmic reticulum stress, aberrant autophagy, and abnormal post-translational modification of proteins, all of which are signature features of both AD and dysregulated insulin/IGF-1-mechanistic target of rapamycin (mTOR) signaling. This article connects the dots from benign to malignant aging to neurodegeneration by reviewing the salient pathologies associated with initially adaptive and later dysfunctional mTOR signaling in the brain. Effective therapeutic and preventive measures must be two-pronged and designed to 1) address complex and shifting impairments in mTOR signaling through the re-purpose of effective anti-diabetes therapeutics that target the brain, and 2) minimize the impact of extrinsic mediators of benign to malignant aging transitions, e.g., inflammatory states, obesity, systemic insulin resistance diseases, and repeated bouts of general anesthesia, by minimizing exposures or implementing neuroprotective measures.
Collapse
Affiliation(s)
- Suzanne M. de la Monte
- Departments of Pathology and Laboratory Medicine, Medicine, Neurology and Neurosurgery, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
18
|
Kopp KO, Glotfelty EJ, Li Y, Greig NH. Glucagon-like peptide-1 (GLP-1) receptor agonists and neuroinflammation: Implications for neurodegenerative disease treatment. Pharmacol Res 2022; 186:106550. [PMID: 36372278 PMCID: PMC9712272 DOI: 10.1016/j.phrs.2022.106550] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/03/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2022]
Abstract
Chronic, excessive neuroinflammation is a key feature of neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD). However, neuroinflammatory pathways have yet to be effectively targeted in clinical treatments for such diseases. Interestingly, increased inflammation and neurodegenerative disease risk have been associated with type 2 diabetes mellitus (T2DM) and insulin resistance (IR), suggesting that treatments that mitigate T2DM pathology may be successful in treating neuroinflammatory and neurodegenerative pathology as well. Glucagon-like peptide-1 (GLP-1) is an incretin hormone that promotes healthy insulin signaling, regulates blood sugar levels, and suppresses appetite. Consequently, numerous GLP-1 receptor (GLP-1R) stimulating drugs have been developed and approved by the US Food and Drug Administration (FDA) and related global regulatory authorities for the treatment of T2DM. Furthermore, GLP-1R stimulating drugs have been associated with anti-inflammatory, neurotrophic, and neuroprotective properties in neurodegenerative disorder preclinical models, and hence hold promise for repurposing as a treatment for neurodegenerative diseases. In this review, we discuss incretin signaling, neuroinflammatory pathways, and the intersections between neuroinflammation, brain IR, and neurodegenerative diseases, with a focus on AD and PD. We additionally overview current FDA-approved incretin receptor stimulating drugs and agents in development, including unimolecular single, dual, and triple receptor agonists, and highlight those in clinical trials for neurodegenerative disease treatment. We propose that repurposing already-approved GLP-1R agonists for the treatment of neurodegenerative diseases may be a safe, efficacious, and cost-effective strategy for ameliorating AD and PD pathology by quelling neuroinflammation.
Collapse
Affiliation(s)
- Katherine O Kopp
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, United States.
| | - Elliot J Glotfelty
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, United States; Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Yazhou Li
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, United States
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, United States.
| |
Collapse
|
19
|
Pačesová A, Holubová M, Hrubá L, Strnadová V, Neprašová B, Pelantová H, Kuzma M, Železná B, Kuneš J, Maletínská L. Age-related metabolic and neurodegenerative changes in SAMP8 mice. Aging (Albany NY) 2022; 14:7300-7327. [PMID: 36126192 PMCID: PMC9550245 DOI: 10.18632/aging.204284] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 08/31/2022] [Indexed: 11/25/2022]
Abstract
The most important risk factor for the development of sporadic Alzheimer's disease (AD) is ageing. Senescence accelerated mouse prone 8 (SAMP8) is a model of sporadic AD, with senescence accelerated resistant mouse (SAMR1) as a control. In this study, we aimed to determine the onset of senescence-induced neurodegeneration and the related potential therapeutic window using behavioral experiments, immunohistochemistry and western blotting in SAMP8 and SAMR1 mice at 3, 6 and 9 months of age. The Y-maze revealed significantly impaired working spatial memory of SAMP8 mice from the 6th month. With ageing, increasing plasma concentrations of proinflammatory cytokines in SAMP8 mice were detected as well as significantly increased astrocytosis in the cortex and microgliosis in the brainstem. Moreover, from the 3rd month, SAMP8 mice displayed a decreased number of neurons and neurogenesis in the hippocampus. From the 6th month, increased pathological phosphorylation of tau protein at Thr231 and Ser214 was observed in the hippocampi of SAMP8 mice. In conclusion, changes specific for neurodegenerative processes were observed between the 3rd and 6th month of age in SAMP8 mice; thus, potential neuroprotective interventions could be applied between these ages.
Collapse
Affiliation(s)
- Andrea Pačesová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 166 10, Czech Republic
| | - Martina Holubová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 166 10, Czech Republic
| | - Lucie Hrubá
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 166 10, Czech Republic
| | - Veronika Strnadová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 166 10, Czech Republic
| | - Barbora Neprašová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 166 10, Czech Republic
| | - Helena Pelantová
- Institute of Microbiology of the Czech Academy of Sciences, Prague 142 00, Czech Republic
| | - Marek Kuzma
- Institute of Microbiology of the Czech Academy of Sciences, Prague 142 00, Czech Republic
| | - Blanka Železná
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 166 10, Czech Republic
| | - Jaroslav Kuneš
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 166 10, Czech Republic
- Institute of Physiology of the Czech Academy of Sciences, Prague 142 00, Czech Republic
| | - Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 166 10, Czech Republic
| |
Collapse
|
20
|
Xu Z, Zhang J, Wu J, Yang S, Li Y, Wu Y, Li S, Zhang X, Zuo W, Lian X, Lin J, Jiang Y, Xie L, Liu Y, Wang P. Lactobacillus plantarum ST-III culture supernatant ameliorates alcohol-induced cognitive dysfunction by reducing endoplasmic reticulum stress and oxidative stress. Front Neurosci 2022; 16:976358. [PMID: 36188464 PMCID: PMC9515438 DOI: 10.3389/fnins.2022.976358] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/15/2022] [Indexed: 11/28/2022] Open
Abstract
Background Long-term alcohol exposure is associated with oxidative stress, endoplasmic reticulum (ER) stress, and neuroinflammation, which may impair cognitive function. Probiotics supplements can significantly improve cognitive function in neurodegenerative diseases such as Alzheimer’s disease. Nevertheless, the effect of Lactobacillus plantarum ST-III culture supernatant (LP-cs) on alcohol-induced cognitive dysfunction remains unclear. Methods A mouse model of cognitive dysfunction was established by intraperitoneal injection of alcohol (2 g/kg body weight) for 28 days. Mice were pre-treated with LP-cs, and cognitive function was evaluated using the Morris water maze test. Hippocampal tissues were collected for biochemical and molecular analysis. Results LP-cs significantly ameliorated alcohol-induced decline in learning and memory function and hippocampal morphology changes, neuronal apoptosis, and synaptic dysfunction. A mechanistic study showed that alcohol activated protein kinase R-like endoplasmic reticulum kinase (PERK) signaling and suppressed brain derived neurotrophic factor (BDNF) levels via ER stress in the hippocampus, which LP-cs reversed. Alcohol activated oxidative stress and inflammation responses in the hippocampus, which LP-cs reversed. Conclusion LP-cs significantly ameliorated alcohol-induced cognitive dysfunction and cellular stress. LP-cs might serve as an effective treatment for alcohol-induced cognitive dysfunction.
Collapse
Affiliation(s)
- Zeping Xu
- Department of Pharmacy, Ningbo Medical Center Li Huili Hospital, The Affiliated Hospital of Ningbo University, Ningbo, China
| | - Jinjing Zhang
- Department of Pharmacy, Affiliated Cixi Hospital, Wenzhou Medical University, Wenzhou, China
| | - Junnan Wu
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
- The Affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou, China
| | - Shizhuo Yang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yuying Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yuyu Wu
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Siyuan Li
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Xie Zhang
- Department of Pharmacy, Ningbo Medical Center Li Huili Hospital, The Affiliated Hospital of Ningbo University, Ningbo, China
| | - Wei Zuo
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, China
| | - Xiang Lian
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, China
| | - Jianjun Lin
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, China
| | - Yongsheng Jiang
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, China
| | - Longteng Xie
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, China
- Longteng Xie,
| | - Yanlong Liu
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
- The Affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Yanlong Liu,
| | - Ping Wang
- The Affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Ping Wang,
| |
Collapse
|