1
|
Alzarea EA, Al-Kuraishy HM, Al-Gareeb AI, Alexiou A, Papadakis M, Beshay ON, Batiha GES. The Conceivable Role of Metabolic Syndrome in the Pathogenesis of Alzheimer's Disease: Cellular and Subcellular Alterations in Underpinning a Tale of Two. Neuromolecular Med 2025; 27:35. [PMID: 40379890 DOI: 10.1007/s12017-025-08832-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/09/2025] [Indexed: 05/19/2025]
Abstract
Alzheimer's disease (AD)is an age-related neurodegenerative disease characterized by memory decline and cognitive impairment .AD is common in people aged > 65 years, though most of AD cases are sporadic, which accounts for 95%, and 1-5% of AD is caused by familial causes . The causes of AD are aging, environmental toxins, and cardiometabolic factors that induce the degeneration of cholinergic neurons. It has been shown that the metabolic syndrome which is a clustering of dissimilar constituents including insulin resistance (IR), glucose intolerance, visceral obesity, hypertension, and dyslipidemia is implicated in the pathogenesis of AD. Metabolic syndrome disapprovingly affects cognitive function and the development in AD by inducing the development of oxidative stress, neuroinflammation, and brain IR. These changes, together with brain IR, impair cerebrovascular reactivity causing cognitive impairment and dementia. Nevertheless, the fundamental mechanism by which metabolic syndrome persuades AD risk is not entirely explicated. Accordingly, this review aims to discuss the connotation between metabolic syndrome and AD. In conclusion, metabolic syndrome is regarded as a possible risk factor for the initiation of AD neuropathology by diverse signaling pathways such as brain IR, activation of inflammatory signaling pathways, neuroinflammation, defective proteostasis, and dysregulation of lipid mediators.
Collapse
Affiliation(s)
- Ekremah A Alzarea
- Hematopathology, Department of Pathology, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Jabir Ibn Hayyan Medical University, Al-Ameer Qu./Najaf-Iraq, PO.Box13, Kufa, Iraq
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, Australia
- Department of Research & Development, Funogen, Athens, Greece
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Olivia N Beshay
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt
| |
Collapse
|
2
|
Grabowska K, Grabowski M, Burek M, Meybohm P, Przybyła M, Barski JJ, Małecki A, Nowacka-Chmielewska M. Exercise Training Alters the Hippocampal Expression of Blood-Brain Barrier Components and Behavior of Western Diet-Fed Female Rats. Mol Neurobiol 2025:10.1007/s12035-025-04873-x. [PMID: 40164886 DOI: 10.1007/s12035-025-04873-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Overeating highly palatable foods typical of a Western diet (WD) has been linked to cognitive impairments in animal models and humans. Exercise training was proposed as an important behavioral intervention with beneficial effects, including improving peripheral insulin sensitivity, improving central functions such as learning and memory, and restoring a dysregulated blood-brain barrier (BBB). The purpose of the present study was to characterize the effect of exercise training in rats fed with the WD with special emphasis on BBB. Adult female Long Evans rats were subjected to 12 weeks of WD feeding (WD group), or simultaneous WD feeding and wheel-running training (WD/EX group), or were fed a WD for 6 weeks without training and then subjected to diet and training for an additional 6 weeks (WD_WD/EX group). A sedentary (untrained) group of lean rats was fed a standard rodent chow (CTR group). In all experimental groups, we measured behavioral and physiological parameters, and the hippocampal levels of proteins structurally and functionally related to BBB, including proinflammatory cytokines and products of elevated lipid peroxidation. Exercise training in combination with a WD decreased locomotor and exploratory activities and induced short-term memory impairments. The behavioral changes were accompanied by reduced levels of occludin, claudin-5, and ZO-1 proteins in the hippocampus, suggesting changes in the integrity and increased permeability of BBB. In the WD_WD/EX rats, we found increased hippocampal levels of malondialdehyde (MDA) and neurotrophins (Bdnf, Vegfa) suggesting that increased energy expenditure by obese rats stimulates endogenous protective processes. The training introduced after 6 weeks of WD feeding in rats showing an obese phenotype may suggest that the sequence and moment of presumably protective intervention (exercise training) could alleviate or, on the contrary, exacerbate the level of stress and its consequences.
Collapse
Affiliation(s)
- Konstancja Grabowska
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, Academy of Physical Education, Katowice, Poland
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Mateusz Grabowski
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, Academy of Physical Education, Katowice, Poland
| | - Małgorzata Burek
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Patrick Meybohm
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Marta Przybyła
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, Academy of Physical Education, Katowice, Poland
- Department for Experimental Medicine, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Jarosław J Barski
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Andrzej Małecki
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, Academy of Physical Education, Katowice, Poland
| | - Marta Nowacka-Chmielewska
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, Academy of Physical Education, Katowice, Poland.
| |
Collapse
|
3
|
Wang J, Liao M, Tong Z, Yuan S, Hu Z, Chen Z, Zeng F, Zou R, Chen D, Chen G, Wang Z, Liu W. Treadmill Exercise Modulates the Leptin/LepR/GSK-3β Signalling Pathway to Improve Leptin Sensitivity and Alleviate Neuroinflammation in High-Fat Diet-Fed APP/PS1 Mice. Mol Neurobiol 2025:10.1007/s12035-025-04853-1. [PMID: 40131695 DOI: 10.1007/s12035-025-04853-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 03/12/2025] [Indexed: 03/27/2025]
Abstract
Neuroinflammation plays a critical role in the development of Alzheimer's disease (AD) and is closely associated with obesity. In AD, the fat cell-secreted protein leptin crosses the blood-brain barrier and protects against nerve damage. However, obesity may induce leptin resistance, reduce leptin sensitivity, stimulate excessive glial cell activation, promote inflammatory factor production and exacerbate brain inflammation. Unfortunately, the mechanism of interaction among high-fat diets, obesity, neuroinflammation and neurodegenerative diseases remains unclear. We investigated the changes in neuroinflammation and leptin sensitivity in the brains of wild-type and high-fat-diet-fed APP/PS1 transgenic mice. We explored the effects of treadmill exercise for 12 weeks on the leptin/LepR/GSK-3β signalling pathway and memory. The body weights of the high-fat-diet-fed mice increased, and elevated levels of markers for leptin resistance, including suppressor of signalling 3 (SOCS3), protein tyrosine phosphatase 1B (PTP1B) and proinflammatory factors such as tumour necrosis factor-α (TNF-α) and interleukin-6 (IL-6), were observed. After 12 weeks of aerobic exercise, the leptin mRNA and protein levels increased, GSK-3β protein expression decreased and the mean fluorescence intensities of brain microglial (IBA-1) and neuron markers (NeuN) decreased, indicating that exercise may activate the leptin/LepR/GSK-3β signalling pathway, reducing glial cell activation and inflammation. Our study revealed that obesity induces and exacerbates the AD-related neuroinflammatory response. Aerobic exercise activates the leptin/LepR/GSK-3β pathway to relieve neuroinflammation and protect nerve cells, alleviating AD-associated memory loss. These promising outcomes could inform the development of nondrug-based aerobic exercise interventions for the treatment of AD and associated cognitive disorders.
Collapse
Affiliation(s)
- Juan Wang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Meiqing Liao
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Zhen Tong
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Shunling Yuan
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Zelin Hu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Zeyu Chen
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Fanqi Zeng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Ruihan Zou
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Dandan Chen
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Gan Chen
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China.
- Key Laboratory of Protein Chemistry and Developmental Biology, Ministry of Education, Physical Education College, Hunan Normal University, Yuelu District, No. 437, Lushan South Road, Changsha, 410081, China.
| | - Zhiyuan Wang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China.
- Key Laboratory of Protein Chemistry and Developmental Biology, Ministry of Education, Physical Education College, Hunan Normal University, Yuelu District, No. 437, Lushan South Road, Changsha, 410081, China.
| | - Wenfeng Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China.
- Key Laboratory of Protein Chemistry and Developmental Biology, Ministry of Education, Physical Education College, Hunan Normal University, Yuelu District, No. 437, Lushan South Road, Changsha, 410081, China.
| |
Collapse
|
4
|
Ali NH, Al‐Kuraishy HM, Al‐Gareeb AI, Alexiou A, Papadakis M, Bahaa MM, Alibrahim F, Batiha GE. New insight on the potential detrimental effect of metabolic syndrome on the Alzheimer disease neuropathology: Mechanistic role. J Cell Mol Med 2024; 28:e70118. [PMID: 39644152 PMCID: PMC11624485 DOI: 10.1111/jcmm.70118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 08/29/2024] [Accepted: 09/13/2024] [Indexed: 12/09/2024] Open
Abstract
The metabolic syndrome or syndrome X is a clustering of different components counting insulin resistance (IR), glucose intolerance, visceral obesity, hypertension and dyslipidemia. It has been shown that IR and dysregulation of insulin signalling play a critical role in the development of metabolic syndrome by initiating the pathophysiology of metabolic syndrome through induction of glucolipotoxicity, impairment of glucose disposal and triggering of pro-inflammatory response. Furthermore, metabolic syndrome unfavourably affects the cognitive function and the development of different neurodegenerative diseases such as Alzheimer disease (AD) by inducing oxidative stress, neuroinflammation and brain IR. These changes together with brain IR impair cerebrovascular reactivity leading to cognitive impairment. In addition, metabolic syndrome increases the risk for the development of AD. However, the central mechanisms by which metabolic syndrome amplify AD risk are not completely elucidated. Consequently, this narrative review aims to revise from published articles the association between metabolic syndrome and AD regarding cellular and subcellular pathways. In conclusion, metabolic syndrome is regarded as a potential risk factor for the induction of AD neuropathology by different signalling pathways such as initiation of brain IR, activation of inflammatory signalling pathways and neuroinflammation.
Collapse
Affiliation(s)
- Naif H. Ali
- Department of Internal Medicine, Medical CollegeNajran UniversityNajranSaudi Arabia
| | - Hayder M. Al‐Kuraishy
- Department of Clinical pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | | | - Athanasios Alexiou
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
- Department of Research & DevelopmentAFNP MedWienAustria
- Department of Research & DevelopmentFunogenAthensGreece
- University Centre for Research & DevelopmentChandigarh UniversityMohaliPunjabIndia
| | - Marios Papadakis
- Department of Surgery IIUniversity Hospital Witten‐Herdecke, University of Witten‐HerdeckeWuppertalGermany
| | - Mostafa M. Bahaa
- Pharmacy Practice Department, Faculty of PharmacyHorus UniversityNew DamiettaEgypt
| | - Fawaz Alibrahim
- Division of NeurologyKing Abdulaziz Medical City, Ministry of the National Guard Health AffairsRiyadhSaudi Arabia
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourEgypt
| |
Collapse
|
5
|
Faldu KG, Shah JS. Ambroxol Improves Amyloidogenic, NF-κB, and Nrf2 Pathways in a Scopolamine-Induced Cognitive Impairment Rat Model of Alzheimer's Disease. Drug Dev Res 2024; 85:e70017. [PMID: 39533780 DOI: 10.1002/ddr.70017] [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: 06/17/2024] [Revised: 10/13/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Ambroxol (ABX) is used to manage excessive production of mucus in the respiratory system. The present study sought to assess the neuroprotective potential of ambroxol by influencing the amyloidogenic, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), and nuclear factor erythroid 2-related factor 2 (Nrf2) pathways in a rat model of Alzheimer's disease (AD) induced by scopolamine. The AD pathology was induced by chronic administration of scopolamine. The rats were given scopolamine at a dose of 2 mg/kg via intraperitoneal injection daily for 14 days, followed by treatment (ABX 121.5, 135, and 180 mg/kg orally and 5 mg/kg orally of donepezil) for the next 28 days while continuing to receive daily scopolamine injection. The behavior of the rats was evaluated using Modified Y-Maze and Novel object recognition tasks. Analyses were carried out on AD pathological markers [Amyloid beta peptide 1-40, Amyloid beta peptide 1-42, acetylcholinesterase, beta-secretase 1 (BACE1), total tau, and p-tau], inflammatory markers [NF-κB, tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6), and interferon γ], antioxidant markers (Nrf2 and heme Oxygenase 1 (HO-1)], along with synaptophysin and glial fibrillary acidic protein (GFAP) immunohistochemistry and histopathological assessment of the hippocampus. Our findings indicated that ABX reduced impairment in behavior. Levels of Acetylcholinesterase, BACE1, amyloid beta 1-40, amyloid beta 1-42, total tau, p-tau, NF-κB, IFN-γ, IL-6, and TNF-α decreased significantly. There was a significant increase in the levels of HO-1 and Nrf2. It stopped the neuronal degeneration, raised synaptophysin immunoreactivity, and lowered GFAP immunoreactivity. The current research indicates that ambroxol may possess senomorphic properties by impacting the transcription factors NF-κB and senescence-associated secretory phenotype (SASP). Consequently, it could provide neuroprotection through alterations in the Nrf2 and NF-κB signaling pathways in AD.
Collapse
Affiliation(s)
- Khushboo Govind Faldu
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Jigna Samir Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| |
Collapse
|
6
|
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
|
7
|
Ge X, Hu M, Zhou M, Fang X, Chen X, Geng D, Wang L, Yang X, An H, Zhang M, Lin D, Zheng M, Cui X, Wang Q, Wu Y, Zheng K, Huang XF, Yu Y. Overexpression of forebrain PTP1B leads to synaptic and cognitive impairments in obesity. Brain Behav Immun 2024; 117:456-470. [PMID: 38336024 DOI: 10.1016/j.bbi.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/23/2024] [Accepted: 02/04/2024] [Indexed: 02/12/2024] Open
Abstract
Obesity has reached pandemic proportions and is a risk factor for neurodegenerative diseases, including Alzheimer's disease. Chronic inflammation is common in obese patients, but the mechanism between inflammation and cognitive impairment in obesity remains unclear. Accumulative evidence shows that protein-tyrosine phosphatase 1B (PTP1B), a neuroinflammatory and negative synaptic regulator, is involved in the pathogenesis of neurodegenerative processes. We investigated the causal role of PTP1B in obesity-induced cognitive impairment and the beneficial effect of PTP1B inhibitors in counteracting impairments of cognition, neural morphology, and signaling. We showed that obese individuals had negative relationship between serum PTP1B levels and cognitive function. Furthermore, the PTP1B level in the forebrain increased in patients with neurodegenerative diseases and obese cognitive impairment mice with the expansion of white matter, neuroinflammation and brain atrophy. PTP1B globally or forebrain-specific knockout mice on an obesogenic high-fat diet showed enhanced cognition and improved synaptic ultrastructure and proteins in the forebrain. Specifically, deleting PTP1B in leptin receptor-expressing cells improved leptin synaptic signaling and increased BDNF expression in the forebrain of obese mice. Importantly, we found that various PTP1B allosteric inhibitors (e.g., MSI-1436, well-tolerated in Phase 1 and 1b clinical trials for obesity and type II diabetes) prevented these alterations, including improving cognition, neurite outgrowth, leptin synaptic signaling and BDNF in both obese cognitive impairment mice and a neural cell model of PTP1B overexpression. These findings suggest that increased forebrain PTP1B is associated with cognitive decline in obesity, whereas inhibition of PTP1B could be a promising strategy for preventing neurodegeneration induced by obesity.
Collapse
Affiliation(s)
- Xing Ge
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Minmin Hu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Menglu Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xiaoli Fang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu 221006, China
| | - Xi Chen
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medical, Indigenous, and Health, University of Wollongong, NSW 2522, Australia
| | - Deqin Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu 221006, China
| | - Li Wang
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110032, China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Huimei An
- HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Peking University, Beijing 10096, China
| | - Meng Zhang
- HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Peking University, Beijing 10096, China
| | - Danhong Lin
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Mingxuan Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xiaoying Cui
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4113, Australia; Queensland Centre for Mental Health Research, Wacol, QLD, 4076, Australia
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Yuqing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China.
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| | - Xu-Feng Huang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Illawarra Health and Medical Research Institute (IHMRI) and School of Medical, Indigenous, and Health, University of Wollongong, NSW 2522, Australia.
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
8
|
Liemisa B, Newbury SF, Novy MJ, Pasato JA, Morales-Corraliza J, Peng KY, Mathews PM. Brain apolipoprotein E levels in mice challenged by a Western diet increase in an allele-dependent manner. AGING BRAIN 2023; 4:100102. [PMID: 38058491 PMCID: PMC10696459 DOI: 10.1016/j.nbas.2023.100102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/05/2023] [Accepted: 11/17/2023] [Indexed: 12/08/2023] Open
Abstract
Human apolipoprotein E (APOE) is the greatest determinant of genetic risk for memory deficits and Alzheimer's disease (AD). While APOE4 drives memory loss and high AD risk, APOE2 leads to healthy brain aging and reduced AD risk compared to the common APOE3 variant. We examined brain APOE protein levels in humanized mice homozygous for these alleles and found baseline levels to be age- and isoform-dependent: APOE2 levels were greater than APOE3, which were greater than APOE4. Despite the understanding that APOE lipoparticles do not traverse the blood-brain barrier, we show that brain APOE levels are responsive to dietary fat intake. Challenging mice for 6 months on a Western diet high in fat and cholesterol increased APOE protein levels in an allele-dependent fashion with a much greater increase within blood plasma than within the brain. In the brain, APOE2 levels responded most to the Western diet challenge, increasing by 20 % to 30 %. While increased lipoparticles are generally deleterious in the periphery, we propose that higher brain APOE2 levels may represent a readily available pool of beneficial lipid particles for neurons.
Collapse
Affiliation(s)
- Braison Liemisa
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Samantha F. Newbury
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Mariah J. Novy
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Jonathan A. Pasato
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Jose Morales-Corraliza
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Katherine Y. Peng
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Paul M. Mathews
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
- NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
9
|
García-García I, Donica O, Cohen AA, Gonseth Nusslé S, Heini A, Nusslé S, Pichard C, Rietschel E, Tanackovic G, Folli S, Draganski B. Maintaining brain health across the lifespan. Neurosci Biobehav Rev 2023; 153:105365. [PMID: 37604360 DOI: 10.1016/j.neubiorev.2023.105365] [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: 03/14/2023] [Revised: 07/24/2023] [Accepted: 08/17/2023] [Indexed: 08/23/2023]
Abstract
Across the lifespan, the human body and brain endure the impact of a plethora of exogenous and endogenous factors that determine the health outcome in old age. The overwhelming inter-individual variance spans between progressive frailty with loss of autonomy to largely preserved physical, cognitive, and social functions. Understanding the mechanisms underlying the diverse aging trajectories can inform future strategies to maintain a healthy body and brain. Here we provide a comprehensive overview of the current literature on lifetime factors governing brain health. We present the growing body of evidence that unhealthy alimentary regime, sedentary behaviour, sleep pathologies, cardio-vascular risk factors, and chronic inflammation exert their harmful effects in a cumulative and gradual manner, and that timely and efficient intervention could promote healthy and successful aging. We discuss the main effects and interactions between these risk factors and the resulting brain health outcomes to follow with a description of current strategies aiming to eliminate, treat, or counteract the risk factors. We conclude that the detailed insights about modifiable risk factors could inform personalized multi-domain strategies for brain health maintenance on the background of increased longevity.
Collapse
Affiliation(s)
- Isabel García-García
- Laboratory for Research in Neuroimaging (LREN), Department of Clinical Neurosciences, Centre for Research in Neurosciences, Lausanne University Hospital, University of Lausanne, Switzerland; Clinique la Prairie, Montreux, Switzerland
| | | | - Armand Aaron Cohen
- Department of Geriatrics and Rehabilitation, Hadassah University Medical Center Mount Scopus, Jerusalem, Israel
| | | | | | | | - Claude Pichard
- Nutrition Unit, University Hospital of Geneva, Geneva, Switzerland
| | | | | | | | - Bogdan Draganski
- Laboratory for Research in Neuroimaging (LREN), Department of Clinical Neurosciences, Centre for Research in Neurosciences, Lausanne University Hospital, University of Lausanne, Switzerland; Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
| |
Collapse
|
10
|
Alves SS, Servilha-Menezes G, Rossi L, da Silva Junior RMP, Garcia-Cairasco N. Evidence of disturbed insulin signaling in animal models of Alzheimer's disease. Neurosci Biobehav Rev 2023; 152:105326. [PMID: 37479008 DOI: 10.1016/j.neubiorev.2023.105326] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 06/02/2023] [Accepted: 07/17/2023] [Indexed: 07/23/2023]
Abstract
Since glucose reuptake by neurons is mostly independent of insulin, it has been an intriguing question whether insulin has or not any roles in the brain. Consequently, the identification of insulin receptors in the central nervous system has fueled investigations of insulin functions in the brain. It is also already known that insulin can influence glucose reuptake by neurons, mostly during activities that have the highest energy demand. The identification of high density of insulin receptors in the hippocampus also suggests that insulin may present important roles related to memory. In this context, studies have reported worse performance in cognitive tests among diabetic patients. In addition, alterations in the regulation of central insulin pathways have been observed in the brains of Alzheimer's disease (AD) patients. In fact, some authors have proposed AD as a third type of diabetes and recently, our group proposed insulin resistance as a common link between different AD hypotheses. Therefore, in the present narrative review, we intend to revise and gather the evidence of disturbed insulin signaling in experimental animal models of AD.
Collapse
Affiliation(s)
- Suélen Santos Alves
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo (FMRP-USP), Brazil
| | - Gabriel Servilha-Menezes
- Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Brazil
| | - Leticia Rossi
- Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Brazil
| | - Rui Milton Patrício da Silva Junior
- Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Brazil; Institute of Neuroscience of Castilla y León, University of Salamanca, Salamanca, Spain
| | - Norberto Garcia-Cairasco
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo (FMRP-USP), Brazil; Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Brazil.
| |
Collapse
|
11
|
Rosca CI, Lighezan DF, Nisulescu DD, Sharma A, Neagu MN, Nistor D, Georgescu D, Kundnani NR. Metabolic Syndrome: A Strange Companion of Atrial Fibrillation; A Blessing in Disguise from the Neuropsychiatric Point of View. Biomedicines 2023; 11:2012. [PMID: 37509651 PMCID: PMC10377522 DOI: 10.3390/biomedicines11072012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/12/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Background: The concept of metabolic syndrome (MetSy) brings together components that individually represent a risk factor for cardiovascular diseases, which over time can prove to be more harmful if a combined effect of these is exhibited. Method: A single-centre retrospective study in an academic medical unit was conducted. We analysed the link between the MetSy and the occurrence of neuropsychic complications among atrial fibrillation (AF) patients. We sifted through the files of the patients admitted during 2015-2016 to the Municipal Emergency University Hospital Timisoara, Romania, with the diagnosis of AF. We divided these AF patients into two groups: the first group comprised patients with atrial fibrillation and MetSy (267 patients), while the second group comprised AF patients without MetSy (843 patients). We analysed the occurrence of neuropsychic changes (stroke, Parkinson's disease, dementia, cognitive impairment, and silent lacunar infarction) among the two groups. Results: Cognitive impairment (p-value = 0.0081) and dementia (p-value < 0.0001) were less frequent in patients with AF and MetSy than in those with AF without MetSy. Regarding the presence of stroke and Parkinson's disease (PD), we could not demonstrate the existence of any statistically significant difference between the two groups. Using logistic regression (enter test), we found that MetSy might have a protective effect (OR = 0.4040, 95% CI [0.2132; 0.7654], p-value = 0.0054) for the occurrence of dementia in those patients. Furthermore, obesity was the only factor with a possible protective effect from all the constituents of the MetSy when analysed together (with a significance level of p-value = 0.0004 for the logistic regression). The protective effect of MetSy against stroke occurrence was supplementarily proven by a longer period of survival without stroke from the AF diagnosis (3.521 years, p = 0.0304) compared to patients with AF without MetSy (3.286 years to first stroke occurrence). Conclusions: Metabolic syndrome might offer protection against the occurrence of dementia among patients with AF, but no effect was noted when compared with the presence of stroke. Further studies on larger cohorts can help us reach a conclusion regarding the positive effects of the metabolic syndrome.
Collapse
Affiliation(s)
- Ciprian Ilie Rosca
- Department of Internal Medicine I-Medical Semiotics I, Centre for Advanced Research in Cardiovascular Pathology and Haemostasis, "Victor Babeș" University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania
| | - Daniel Florin Lighezan
- Department of Internal Medicine I-Medical Semiotics I, Centre for Advanced Research in Cardiovascular Pathology and Haemostasis, "Victor Babeș" University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania
| | | | - Abhinav Sharma
- Department of Cardiology-Internal Medicine and Ambulatory Care, Prevention and Cardiovascular Recovery, "Victor Babeș" University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 3000041 Timișoara, Romania
| | - Marioara Nicula Neagu
- Faculty of Bioengineering of Animal Resources, Discipline of Physiology, University of Life Sciences "King Mihai I" from Timisoara, 300645 Timisoara, Romania
| | - Daciana Nistor
- Department of Functional Sciences, Physiology, Centre of Imuno-Physiology and Biotechnologies (CIFBIOTEH), Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Centre for Gene and Cellular Therapies in Cancer, 3000723 Timisoara, Romania
| | - Doina Georgescu
- Department of Internal Medicine I-Medical Semiotics I, Centre for Advanced Research in Cardiovascular Pathology and Haemostasis, "Victor Babeș" University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 300041 Timișoara, Romania
| | - Nilima Rajpal Kundnani
- Department of Cardiology-Internal Medicine and Ambulatory Care, Prevention and Cardiovascular Recovery, "Victor Babeș" University of Medicine and Pharmacy, Eftimie Murgu Sq. no. 2, 3000041 Timișoara, Romania
| |
Collapse
|
12
|
Zhang Z, Guo Z, Tu Z, Yang H, Li C, Hu M, Zhang Y, Jin P, Hou S. Cortex-specific transcriptome profiling reveals upregulation of interferon-regulated genes after deeper cerebral hypoperfusion in mice. Front Physiol 2023; 14:1056354. [PMID: 36994418 PMCID: PMC10040763 DOI: 10.3389/fphys.2023.1056354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
Background: Chronic cerebral hypoperfusion (CCH) is commonly accompanied by brain injury and glial activation. In addition to white matter lesions, the intensity of CCH greatly affects the degree of gray matter damage. However, little is understood about the underlying molecular mechanisms related to cortical lesions and glial activation following hypoperfusion. Efforts to investigate the relationship between neuropathological alternations and gene expression changes support a role for identifying novel molecular pathways by transcriptomic mechanisms.Methods: Chronic cerebral ischemic injury model was induced by the bilateral carotid artery stenosis (BCAS) using 0.16/0.18 mm microcoils. Cerebral blood flow (CBF) was evaluated using laser speckle contrast imaging (LSCI) system. Spatial learning and memory were assessed by Morris water maze test. Histological changes were evaluated by Hematoxylin staining. Microglial activation and neuronal loss were further examined by immunofluorescence staining. Cortex-specific gene expression profiling analysis was performed in sham and BCAS mice, and then validated by quantitative RT-PCR and immunohistochemistry (IHC).Results: In our study, compared with the sham group, the right hemisphere CBF of BCAS mice decreased to 69% and the cognitive function became impaired at 4 weeks postoperation. Besides, the BCAS mice displayed profound gray matter damage, including atrophy and thinning of the cortex, accompanied by neuronal loss and increased activated microglia. Gene set enrichment analysis (GSEA) revealed that hypoperfusion-induced upregulated genes were significantly enriched in the pathways of interferon (IFN)-regulated signaling along with neuroinflammation signaling. Ingenuity pathway analysis (IPA) predicted the importance of type I IFN signaling in regulating the CCH gene network. The obtained RNA-seq data were validated by qRT-PCR in cerebral cortex, showing consistency with the RNA-seq results. Also, IHC staining revealed elevated expression of IFN-inducible protein in cerebral cortex following BCAS-hypoperfusion.Conclusion: Overall, the activation of IFN-mediated signaling enhanced our understanding of the neuroimmune responses induced by CCH. The upregulation of IFN-regulated genes (IRGs) might exert a critical impact on the progression of cerebral hypoperfusion. Our improved understanding of cortex-specific transcriptional profiles will be helpful to explore potential targets for CCH.
Collapse
Affiliation(s)
- Zengyu Zhang
- Department of Neurology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Zimin Guo
- Department of Neurology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Zhilan Tu
- Department of Neurology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Hualan Yang
- Department of Neurology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Chao Li
- School of Pharmacy, Hubei University of Science and Technology, Hubei, China
| | - Mengting Hu
- Department of Neurology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Yuan Zhang
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Pengpeng Jin
- Department of Chronic Disease Management, Shanghai Pudong Hospital, Fudan University, Shanghai, China
- *Correspondence: Shuangxing Hou, ; Pengpeng Jin,
| | - Shuangxing Hou
- Department of Neurology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
- *Correspondence: Shuangxing Hou, ; Pengpeng Jin,
| |
Collapse
|
13
|
Faldu KG, Patel SS, Shah JS. Celastrus paniculatus oil ameliorates NF-KB mediated neuroinflammation and synaptic plasticity in the scopolamine-induced cognitive impairment rat model. Metab Brain Dis 2023; 38:1405-1419. [PMID: 36809523 DOI: 10.1007/s11011-023-01186-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 02/13/2023] [Indexed: 02/23/2023]
Abstract
BACKGROUND AND AIM Traditionally, Celastrus paniculatus Willd. (CP) oil has been utilized as a tranquilizer and memory enhancer. The present study investigated the neuropharmacological activity and efficacy of CP oil in ameliorating scopolamine-induced cognitive impairment in rats. EXPERIMENTAL PROCEDURE Cognitive deficiency was induced in rats by administration of scopolamine (2 mg/kg intraperitoneal injection) for a period of 15 days. Donepezil served as a reference drug and CP oil was tested as both preventive and curative treatments. Animals' behaviour was assessed through the Morris water maze (MWM), novel object preference (NOR), and conditioned avoidance (CA) tests. Oxidative stress parameters, bioamine concentration (dopamine, noradrenaline, and 5-hydroxytryptamine), nerve growth factor (NGF), interleukin-6 (IL-6), nuclear factor kappa B (NF-кB), and tumor necrosis factor-alpha (TNFα) were estimated. Synaptophysin immunohistochemistry was performed. RESULTS Our results showed that CP oil ameliorated behavioural deficits. It reduced latency to find a hidden platform in MWM. Reduced novel object exploration time and discrimination index (p < 0.05) in the NOR. Reduced step-down latency and normalized conditioned avoidance response (p < 0.001) in the CA test. CP oil increased dopamine, serotonin, norepinephrine, superoxide dismutase (SOD), glutathione, and catalase levels. It decreased malondialdehyde (MDA), acetylcholinesterase activity, IL-6, NF-кB (P < 0.001), TNFα, and NGF levels. Treatment showed approximate typical reactivity to synaptophysin. CONCLUSION Our data is suggestive that CP oil treatment improves behavioural test outcomes, increases biogenic amine concentration, and decreases acetylcholinesterase activity, and neuroinflammatory biomarkers. It also restores synaptic plasticity. It thus improves cognitive functions against scopolamine-induced amnesia in rats by improving cholinergic function.
Collapse
Affiliation(s)
- Khushboo Govind Faldu
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Sarkhej - Gandhinagar Hwy, 382481, Gota, Ahmedabad, Gujarat, India
| | | | - Jigna Samir Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Sarkhej - Gandhinagar Hwy, 382481, Gota, Ahmedabad, Gujarat, India.
| |
Collapse
|
14
|
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: 8] [Impact Index Per Article: 4.0] [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
|
15
|
Toledano-Díaz A, Álvarez MI, Toledano A. The relationships between neuroglial and neuronal changes in Alzheimer's disease, and the related controversies II: gliotherapies and multimodal therapy. J Cent Nerv Syst Dis 2022; 14:11795735221123896. [PMID: 36407561 PMCID: PMC9666878 DOI: 10.1177/11795735221123896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 08/05/2022] [Indexed: 08/30/2023] Open
Abstract
Since the original description of Alzheimer´s disease (AD), research into this condition has mainly focused on assessing the alterations to neurons associated with dementia, and those to the circuits in which they are involved. In most of the studies on human brains and in many models of AD, the glial cells accompanying these neurons undergo concomitant alterations that aggravate the course of neurodegeneration. As a result, these changes to neuroglial cells are now included in all the "pathogenic cascades" described in AD. Accordingly, astrogliosis and microgliosis, the main components of neuroinflammation, have been integrated into all the pathogenic theories of this disease, as discussed in this part of the two-part monograph that follows an accompanying article on gliopathogenesis and glioprotection. This initial reflection verified the implication of alterations to the neuroglia in AD, suggesting that these cells may also represent therapeutic targets to prevent neurodegeneration. In this second part of the monograph, we will analyze the possibilities of acting on glial cells to prevent or treat the neurodegeneration that is the hallmark of AD and other pathologies. Evidence of the potential of different pharmacological, non-pharmacological, cell and gene therapies (widely treated) to prevent or treat this disease is now forthcoming, in most cases as adjuncts to other therapies. A comprehensive AD multimodal therapy is proposed in which neuronal and neuroglial pharmacological treatments are jointly considered, as well as the use of new cell and gene therapies and non-pharmacological therapies that tend to slow down the progress of dementia.
Collapse
|
16
|
Lee D, Lee VMY, Hur SK. Manipulation of the diet-microbiota-brain axis in Alzheimer's disease. Front Neurosci 2022; 16:1042865. [PMID: 36408394 PMCID: PMC9672822 DOI: 10.3389/fnins.2022.1042865] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Several studies investigating the pathogenesis of Alzheimer's disease have identified various interdependent constituents contributing to the exacerbation of the disease, including Aβ plaque formation, tau protein hyperphosphorylation, neurofibrillary tangle accumulation, glial inflammation, and the eventual loss of proper neural plasticity. Recently, using various models and human patients, another key factor has been established as an influential determinant in brain homeostasis: the gut-brain axis. The implications of a rapidly aging population and the absence of a definitive cure for Alzheimer's disease have prompted a search for non-pharmaceutical tools, of which gut-modulatory therapies targeting the gut-brain axis have shown promise. Yet multiple recent studies examining changes in human gut flora in response to various probiotics and environmental factors are limited and difficult to generalize; whether the state of the gut microbiota in Alzheimer's disease is a cause of the disease, a result of the disease, or both through numerous feedback loops in the gut-brain axis, remains unclear. However, preliminary findings of longitudinal studies conducted over the past decades have highlighted dietary interventions, especially Mediterranean diets, as preventative measures for Alzheimer's disease by reversing neuroinflammation, modifying the intestinal and blood-brain barrier (BBB), and addressing gut dysbiosis. Conversely, the consumption of Western diets intensifies the progression of Alzheimer's disease through genetic alterations, impaired barrier function, and chronic inflammation. This review aims to support the growing body of experimental and clinical data highlighting specific probiotic strains and particular dietary components in preventing Alzheimer's disease via the gut-brain axis.
Collapse
Affiliation(s)
- Daniel Lee
- Middleton High School, Middleton, WI, United States
| | - Virginia M-Y. Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Seong Kwon Hur
- Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA, United States
| |
Collapse
|
17
|
Fadó R, Molins A, Rojas R, Casals N. Feeding the Brain: Effect of Nutrients on Cognition, Synaptic Function, and AMPA Receptors. Nutrients 2022; 14:nu14194137. [PMID: 36235789 PMCID: PMC9572450 DOI: 10.3390/nu14194137] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022] Open
Abstract
In recent decades, traditional eating habits have been replaced by a more globalized diet, rich in saturated fatty acids and simple sugars. Extensive evidence shows that these dietary factors contribute to cognitive health impairment as well as increase the incidence of metabolic diseases such as obesity and diabetes. However, how these nutrients modulate synaptic function and neuroplasticity is poorly understood. We review the Western, ketogenic, and paleolithic diets for their effects on cognition and correlations with synaptic changes, focusing mainly (but not exclusively) on animal model studies aimed at tracing molecular alterations that may contribute to impaired human cognition. We observe that memory and learning deficits mediated by high-fat/high-sugar diets, even over short exposure times, are associated with reduced arborization, widened synaptic cleft, narrowed post-synaptic zone, and decreased activity-dependent synaptic plasticity in the hippocampus, and also observe that these alterations correlate with deregulation of the AMPA-type glutamate ionotropic receptors (AMPARs) that are crucial to neuroplasticity. Furthermore, we explored which diet-mediated mechanisms modulate synaptic AMPARs and whether certain supplements or nutritional interventions could reverse deleterious effects, contributing to improved learning and memory in older people and patients with Alzheimer’s disease.
Collapse
Affiliation(s)
- Rut Fadó
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, E-08193 Cerdanyola del Vallès, Spain
- Correspondence: ; Tel.: +34-93-504-20-00
| | - Anna Molins
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain
| | - Rocío Rojas
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain
| | - Núria Casals
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| |
Collapse
|
18
|
Chen R, Cai G, Xu S, Sun Q, Luo J, Wang Y, Li M, Lin H, Liu J. Body mass index related to executive function and hippocampal subregion volume in subjective cognitive decline. Front Aging Neurosci 2022; 14:905035. [PMID: 36062154 PMCID: PMC9428252 DOI: 10.3389/fnagi.2022.905035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022] Open
Abstract
Objective This study aims to explore whether body mass index (BMI) level affects the executive function and hippocampal subregion volume of subjective cognitive decline (SCD). Materials and methods A total of 111 participants were included in the analysis, including SCD (38 of normal BMI, 27 of overweight and obesity) and normal cognitive control (NC) (29 of normal BMI, 17 of overweight and obesity). All subjects underwent the Chinese version of the Stroop Color-Word Test (SCWT) to measure the executive function and a high-resolution 3D T1 structural image acquisition. Two-way ANOVA was used to examine the differences in executive function and gray matter volume in hippocampal subregions under different BMI levels between the SCD and NC. Result The subdimensions of executive function in which different BMI levels interact with SCD and NC include inhibition control function [SCWT C-B reaction time(s): F (1,104) = 5.732, p = 0.018], and the hippocampal subregion volume of CA1 [F (1,99) = 8.607, p = 0.004], hippocampal tail [F (1,99) = 4.077, p = 0.046], and molecular layer [F (1,99) = 6.309, p = 0.014]. After correction by Bonferroni method, the population × BMI interaction only had a significant effect on the CA1 (p = 0.004). Further analysis found that the SCWT C-B reaction time of SCD was significantly longer than NC no matter whether it is at the normal BMI level [F (1,104) = 4.325, p = 0.040] or the high BMI level [F (1,104) = 21.530, p < 0.001], and the inhibitory control function of SCD was worse than that of NC. In the normal BMI group, gray matter volume in the hippocampal subregion (CA1) of SCD was significantly smaller than that of NC [F (1,99) = 4.938, p = 0.029]. For patients with SCD, the high BMI group had worse inhibitory control function [F (1,104) = 13.499, p < 0.001] and greater CA1 volume compared with the normal BMI group [F (1,99) = 7.619, p = 0.007]. Conclusion The BMI level is related to the inhibition control function and the gray matter volume of CA1 subregion in SCD. Overweight seems to increase the gray matter volume of CA1 in the elderly with SCD, but it is not enough to compensate for the damage to executive function caused by the disease. These data provide new insights into the relationship between BMI level and executive function of SCD from the perspective of imaging.
Collapse
Affiliation(s)
- Ruilin Chen
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Guiyan Cai
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Shurui Xu
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Qianqian Sun
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jia Luo
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yajun Wang
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Ming Li
- Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Hui Lin
- Department of Physical Education, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jiao Liu
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Rehabilitation Technology, Fuzhou, China
- Traditional Chinese Medicine Rehabilitation Research Center of State Administration of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Key Laboratory of Orthopedics and Traumatology of Traditional Chinese Medicine and Rehabilitation, Ministry of Education, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
19
|
Flores-Cordero JA, Pérez-Pérez A, Jiménez-Cortegana C, Alba G, Flores-Barragán A, Sánchez-Margalet V. Obesity as a Risk Factor for Dementia and Alzheimer's Disease: The Role of Leptin. Int J Mol Sci 2022; 23:5202. [PMID: 35563589 PMCID: PMC9099768 DOI: 10.3390/ijms23095202] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity is a growing worldwide health problem, affecting many people due to excessive saturated fat consumption, lack of exercise, or a sedentary lifestyle. Leptin is an adipokine secreted by adipose tissue that increases in obesity and has central actions not only at the hypothalamic level but also in other regions and nuclei of the central nervous system (CNS) such as the cerebral cortex and hippocampus. These regions express the long form of leptin receptor LepRb, which is the unique leptin receptor capable of transmitting complete leptin signaling, and are the first regions to be affected by chronic neurocognitive deficits, such as mild cognitive impairment (MCI) and Alzheimer's Disease (AD). In this review, we discuss different leptin resistance mechanisms that could be implicated in increasing the risk of developing AD, as leptin resistance is frequently associated with obesity, which is a chronic low-grade inflammatory state, and obesity is considered a risk factor for AD. Key players of leptin resistance are SOCS3, PTP1B, and TCPTP whose signalling is related to inflammation and could be worsened in AD. However, some data are controversial, and it is necessary to further investigate the underlying mechanisms of the AD-causing pathological processes and how altered leptin signalling affects such processes.
Collapse
Affiliation(s)
| | | | | | | | | | - Víctor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology and Immunology, Medical School, Virgen Macarena University Hospital, University of Seville, Av. Sánchez Pizjuan 4, 41009 Sevilla, Spain; (J.A.F.-C.); (A.P.-P.); (C.J.-C.); (G.A.); (A.F.-B.)
| |
Collapse
|
20
|
TLR4 mutation protects neurovascular function and cognitive decline in high-fat diet-fed mice. J Neuroinflammation 2022; 19:104. [PMID: 35488354 PMCID: PMC9052472 DOI: 10.1186/s12974-022-02465-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 04/19/2022] [Indexed: 12/16/2022] Open
Abstract
Background Metabolic syndrome (MS) is defined as a low-grade proinflammatory state in which abnormal metabolic and cardiovascular factors increase the risk of developing cardiovascular disease and neuroinflammation. Events, such as the accumulation of visceral adipose tissue, increased plasma concentrations of free fatty acids, tissue hypoxia, and sympathetic hyperactivity in MS may contribute to the direct or indirect activation of Toll-like receptors (TLRs), specifically TLR4, which is thought to be a major component of this syndrome. Activation of the innate immune response via TLR4 may contribute to this state of chronic inflammation and may be related to the neuroinflammation and neurodegeneration observed in MS. In this study, we investigated the role of TLR4 in the brain microcirculation and in the cognitive performance of high-fat diet (HFD)-induced MS mice. Methods Wild-type (C3H/He) and TLR4 mutant (C3H/HeJ) mice were maintained under a normal diet (ND) or a HFD for 24 weeks. Intravital video-microscopy was used to investigate the functional capillary density, endothelial function, and endothelial–leukocyte interactions in the brain microcirculation. Plasma concentrations of monocyte chemoattractant protein-1 (MCP-1), adipokines and metabolic hormones were measured with a multiplex immunoassay. Brain postsynaptic density protein-95 and synaptophysin were evaluated by western blotting; astrocytic coverage of the vessels, microglial activation and structural capillary density were evaluated by immunohistochemistry. Results The HFD-induced MS model leads to metabolic, hemodynamic, and microcirculatory alterations, as evidenced by capillary rarefaction, increased rolling and leukocyte adhesion in postcapillary venules, endothelial dysfunction, and less coverage of astrocytes in the vessels, which are directly related to cognitive decline and neuroinflammation. The same model of MS reproduced in mice deficient for TLR4 because of a genetic mutation does not generate such changes. Furthermore, the comparison of wild-type mice fed a HFD and a normolipid diet revealed differences in inflammation in the cerebral microcirculation, possibly related to lower TLR4 activation. Conclusions Our results demonstrate that TLR4 is involved in the microvascular dysfunction and neuroinflammation associated with HFD-induced MS and possibly has a causal role in the development of cognitive decline. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02465-3.
Collapse
|
21
|
Al Haj Ahmad RM, Ababneh NA, Al-Domi HA. Brain insulin resistance as a mechanistic mediator links peripheral metabolic disorders with declining cognition. Diabetes Metab Syndr 2022; 16:102468. [PMID: 35364449 DOI: 10.1016/j.dsx.2022.102468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIMS Studies continue to investigate the underlying mechanism of the association between the increased risk of different types of cognitive decline and metabolic dysregulation. Brain insulin resistance (BIR) has been suggested to explain this association. The vital role of insulin in the body has been examined intensively and extensively; however, its role in the brain requires further investigation. Herein, we confined our focus to summarize the role of brain insulin signaling and the negative effect of dysmetabolism on insulin functioning in the brain. METHODS Published scientific manuscripts between 1998 and 2020 that discussed the effect of selected metabolic disorder conditions such as obesity, type 2 diabetes mellitus (T2DM), and high-fat diet (HFD) on brain functions were reviewed. The main keywords used were insulin resistance, brain insulin resistance, obesity, T2DM, and cognition. RESULTS Various metabolic disorders were linked to the increased risk of BIR, and was suggested to increase the probability of cognition impairment occurrence. Several proposed mechanisms explain this association among which insulin resistance and hyperinsulinemia were primary factors attributed to an increased risk of BIR among various metabolic disorders. CONCLUSIONS Understanding the trajectory of the association between metabolic disorders and alternation in cognition status could expand our vision of those overlapping conditions and pave the road to both treatment and preventative strategies for cognitive disorders.
Collapse
Affiliation(s)
- Reem M Al Haj Ahmad
- Department of Nutrition and Food Technology, School of Agriculture, The University of Jordan, Amman, Jordan.
| | - Nidaa A Ababneh
- Cell Therapy Center (CTC), The University of Jordan, Amman, Jordan.
| | - Hayder A Al-Domi
- Department of Nutrition and Food Technology, School of Agriculture, The University of Jordan, Amman, Jordan.
| |
Collapse
|
22
|
MacKenzie JL, Ivanova N, Nell HJ, Giordano CR, Terlecky SR, Agca C, Agca Y, Walton PA, Whitehead SN, Cechetto DF. Microglial inflammation and cognitive dysfunction in comorbid rat models of striatal ischemic stroke and alzheimer’s disease: effects of antioxidant catalase-SKL on behavioral and cellular pathology. Neuroscience 2022; 487:47-65. [DOI: 10.1016/j.neuroscience.2022.01.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 12/25/2022]
|
23
|
Zhou J, Zhang P, Zhang B, Kong Y. White Matter Damage in Alzheimer's Disease: Contribution of Oligodendrocytes. Curr Alzheimer Res 2022; 19:629-640. [PMID: 36281858 PMCID: PMC9982194 DOI: 10.2174/1567205020666221021115321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/15/2022] [Accepted: 09/23/2022] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease seriously influencing the quality of life and is a global health problem. Many factors affect the onset and development of AD, but specific mechanisms underlying the disease are unclear. Most studies investigating AD have focused on neurons and the gray matter in the central nervous system (CNS) but have not led to effective treatments. Recently, an increasing number of studies have focused on white matter (WM). Magnetic resonance imaging and pathology studies have shown different degrees of WM abnormality during the progression of AD. Myelin sheaths, the main component of WM in the CNS, wrap and insulate axons to ensure conduction of the rapid action potential and axonal integrity. WM damage is characterized by progressive degeneration of axons, oligodendrocytes (OLs), and myelin in one or more areas of the CNS. The contributions of OLs to AD progression have, until recently, been largely overlooked. OLs are integral to myelin production, and the proliferation and differentiation of OLs, an early characteristic of AD, provide a promising target for preclinical diagnosis and treatment. However, despite some progress, the key mechanisms underlying the contributions of OLs to AD remain unclear. Given the heavy burden of medical treatment, a better understanding of the pathophysiological mechanisms underlying AD is vital. This review comprehensively summarizes the results on WM abnormalities in AD and explores the relationship between OL progenitor cells and the pathogenesis of AD. Finally, the underlying molecular mechanisms and potential future research directions are discussed.
Collapse
Affiliation(s)
- Jinyu Zhou
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing-400042, China
| | - Peng Zhang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing-400010, China
| | - Bo Zhang
- Department of Basic Medicine, Chongqing Medical and Pharmaceutical College, Chongqing-401331, China
| | - Yuhan Kong
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing-400042, China
| |
Collapse
|
24
|
Li R, Zhan W, Huang X, Zhang L, Zhang Z, Zhou M, Wang Z, Ma Y. The Relationship Between Mild Cognitive Impairment and Anti-Inflammatory/Pro-Inflammatory Nutrients in the Elderly in Northern China: A Bayesian Kernel Machine Regression Approach. J Inflamm Res 2022; 15:325-339. [PMID: 35058706 PMCID: PMC8765544 DOI: 10.2147/jir.s342672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 12/31/2021] [Indexed: 11/23/2022] Open
Affiliation(s)
- Ruiqiang Li
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, People’s Republic of China
| | - Wenqiang Zhan
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Xin Huang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, People’s Republic of China
| | - Limin Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, People’s Republic of China
| | - Zechen Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, People’s Republic of China
| | - Meiqi Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, People’s Republic of China
| | - Zhihong Wang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Yuxia Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, People’s Republic of China
- Correspondence: Yuxia Ma Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang, People’s Republic of China Email
| |
Collapse
|
25
|
Effects of Nutrients on Platelet Function: A Modifiable Link between Metabolic Syndrome and Neurodegeneration? Biomolecules 2021; 11:biom11101455. [PMID: 34680088 PMCID: PMC8533544 DOI: 10.3390/biom11101455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/27/2021] [Accepted: 09/30/2021] [Indexed: 11/17/2022] Open
Abstract
Metabolic syndrome increases the risk of vascular dementia and other neurodegenerative disorders. Recent studies underline that platelets play an important role in linking peripheral with central metabolic and inflammatory mechanisms. In this narrative review, we address the activation of platelets in metabolic syndrome, their effects on neuronal processes and the role of the mediators (e.g., serotonin, platelet-derived growth factor). Emerging evidence shows that nutritional compounds and their metabolites modulate these interactions-specifically, long chain fatty acids, endocannabinoids and phenolic compounds. We reviewed the role of activated platelets in neurovascular processes and nutritional compounds in platelet activation.
Collapse
|
26
|
Urso C, Zhou H. Palmitic Acid Lipotoxicity in Microglia Cells Is Ameliorated by Unsaturated Fatty Acids. Int J Mol Sci 2021; 22:ijms22169093. [PMID: 34445796 PMCID: PMC8396597 DOI: 10.3390/ijms22169093] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/16/2021] [Accepted: 08/20/2021] [Indexed: 01/04/2023] Open
Abstract
Obesity and metabolic syndrome are associated with cognitive decline and dementia. Palmitic acid (PA) is increased in the cerebrospinal fluid of obese patients with cognitive impairment. This study was therefore designed to examine fatty acid (FA) lipotoxicity in BV2 microglia cells. We found that PA induced time- and dose-dependent decrease in cell viability and increase in cell death without affecting the cell cycle profile and that PA lipotoxicity did not depend on cell surface free fatty acid receptors but rather on FA uptake. Treatment with sulfosuccinimidyl oleate (SSO), an irreversible inhibitor of fatty acid translocase CD36, significantly inhibited FA uptake in BSA- and PA-treated cells and blocked PA-induced decrease in cell viability. Inhibition of ER stress or treatment with N-acetylcysteine was not able to rescue PA lipotoxicity. Our study also showed that unsaturated fatty acids (UFAs), such as linoleic acid (LA), oleic acid (OA), α-linolenic acid (ALA), and docosahexaenoic acid (DHA), were not lipotoxic but instead protected microglia against PA-induced decrease in cell viability. Co-treatment of PA with LA, OA, and DHA significantly inhibited FA uptake in PA-treated cells. All UFAs tested induced the incorporation of FAs into and the amount of neutral lipids, while PA did not significantly affect the amount of neutral lipids compared with BSA control.
Collapse
|
27
|
Yin J, Lu J, Lei P, He M, Huang S, Lv J, Zhu Y, Liu Z, Jiang M. Danggui-Shaoyao-San Improves Gut Microbia Dysbiosis and Hepatic Lipid Homeostasis in Fructose-Fed Rats. Front Pharmacol 2021; 12:671708. [PMID: 34326769 PMCID: PMC8313808 DOI: 10.3389/fphar.2021.671708] [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: 02/24/2021] [Accepted: 06/28/2021] [Indexed: 01/02/2023] Open
Abstract
Metabolic syndrome (MetS) is a pathological state of many abnormal metabolic sections. These abnormalities are closely related to diabetes, heart pathologies and other vascular diseases. Danggui-Shaoyao-San (DSS) is a traditional Chinese medicine formula that has been used as a therapy for Alzheimer’s disease. DSS has rarely been reported in the application of MetS and its mechanism of how it improves gut microbia dysbiosis and hepatic lipid homeostasis. In this study, three extracts of DSS were obtained using water, 50% methanol in water and methanol as extracting solvents. Their chemical substances were analyzed by ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass (UPLC-Q/TOF-MS). Pharmacodynamic effect of the extracts were evaluated by comparison of biochemical factors, 16S rRNA sequencing test for gut microbiota analysis, as well as metabonomic and transcriptomic assessments on liver tissues from fructose-fed rats. This study aimed at investigating DSS’s mechanism of regulating blood lipid, anti-inflammation and reducing blood glucose. The results showed that the 50% methanol extract (HME) was more effective. It was worth noting that hydroxysteroid 17β-dehydrogenase 13 (HSD17β13) as a critical element of increasing blood lipid biomarker-triglyceride (TG), was decreased markedly by DSS. The influence from upgraded hydroxysteroid 17β-dehydrogenase 7 (HSD17β7) may be stronger than that from downgraded Lactobacillus in the aspect of regulating back blood lipid biomarker-total cholesterol (TC). The differential down-regulation of tumornecrosis factor alpha (TNF-α) and the significant up-regulation of Akkermansia showed the effective effect of anti-inflammation by DSS. The declining glycine and alanine induced the lowering glucose and lactate. It demonstrated that DSS slowed down the reaction of gluconeogenesis to reduce the blood glucose. The results demonstrated that DSS improved pathological symptoms of MetS and some special biochemical factors in three aspects by better regulating intestinal floras and improving hepatic gene expressions and metabolites.
Collapse
Affiliation(s)
- Jing Yin
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiaxi Lu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Peng Lei
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Mingshuai He
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shengjie Huang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jialin Lv
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yan Zhu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhidong Liu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Miaomiao Jiang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Department of Pharmacy, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
28
|
Joshi C, Jadeja V, Zhou H. Molecular Mechanisms of Palmitic Acid Augmentation in COVID-19 Pathologies. Int J Mol Sci 2021; 22:7127. [PMID: 34281182 PMCID: PMC8269364 DOI: 10.3390/ijms22137127] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 02/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has claimed over 2.7 million lives globally. Obesity has been associated with increased severity and mortality of COVID-19. However, the molecular mechanisms by which obesity exacerbates COVID-19 pathologies are not well-defined. The levels of free fatty acids (FFAs) are elevated in obese subjects. This study was therefore designed to examine how excess levels of different FFAs may affect the progression of COVID-19. Biological molecules associated with palmitic acid (PA) and COVID-19 were retrieved from QIAGEN Knowledge Base, and Ingenuity Pathway Analysis tools were used to analyze these datasets and explore the potential pathways affected by different FFAs. Our study found that one of the top 10 canonical pathways affected by PA was the coronavirus pathogenesis pathway, mediated by key inflammatory mediators, including PTGS2; cytokines, including IL1β and IL6; chemokines, including CCL2 and CCL5; transcription factors, including NFκB; translation regulators, including EEF1A1; and apoptotic mediators, including BAX. In contrast, n-3 fatty acids may attenuate PA's activation of the coronavirus pathogenesis pathway by inhibiting the activity of such mediators as IL1β, CCL2, PTGS2, and BAX. Furthermore, PA may modulate the expression of ACE2, the main cell surface receptor for the SARS-CoV-2 spike protein.
Collapse
Affiliation(s)
| | | | - Heping Zhou
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07079, USA; (C.J.); (V.J.)
| |
Collapse
|
29
|
Alexaki VI. The Impact of Obesity on Microglial Function: Immune, Metabolic and Endocrine Perspectives. Cells 2021; 10:cells10071584. [PMID: 34201844 PMCID: PMC8307603 DOI: 10.3390/cells10071584] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
Increased life expectancy in combination with modern life style and high prevalence of obesity are important risk factors for development of neurodegenerative diseases. Neuroinflammation is a feature of neurodegenerative diseases, and microglia, the innate immune cells of the brain, are central players in it. The present review discusses the effects of obesity, chronic peripheral inflammation and obesity-associated metabolic and endocrine perturbations, including insulin resistance, dyslipidemia and increased glucocorticoid levels, on microglial function.
Collapse
Affiliation(s)
- Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| |
Collapse
|
30
|
Triviño JJ, von Bernhardi R. The effect of aged microglia on synaptic impairment and its relevance in neurodegenerative diseases. Neurochem Int 2021; 144:104982. [PMID: 33556444 DOI: 10.1016/j.neuint.2021.104982] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/31/2021] [Accepted: 02/01/2021] [Indexed: 02/07/2023]
Abstract
Microglia serve key functions in the central nervous system (CNS), participating in the establishment and regulation of synapses and the neuronal network, and regulating activity-dependent plastic changes. As the neuroimmune system, they respond to endogenous and exogenous signals to protect the CNS. In aging, one of the main changes is the establishment of inflamm-aging, a mild chronic inflammation that reduces microglial response to stressors. Neuroinflammation depends mainly on the increased activation of microglia. Microglia over-activation may result in a reduced capacity for performing normal functions related to migration, clearance, and the adoption of an anti-inflammatory state, contributing to an increased susceptibility for neurodegeneration. Oxidative stress contributes both to aging and to the progression of neurodegenerative diseases. Increased production of reactive oxygen species (ROS) and neuroinflammation associated with age- and disease-dependent mechanisms affect synaptic activity and neurotransmission, leading to cognitive dysfunction. Astrocytes prevent microglial cell cytotoxicity by mechanisms mediated by transforming growth factor β1 (TGFβ1). However, TGFβ1-Smad3 pathway is impaired in aging, and the age-related impairment of TGFβ signaling can reduce protective activation while facilitating cytotoxic activation of microglia. A critical analysis on the effect of aging microglia on neuronal function is relevant for the understanding of age-related changes on neuronal function. Here, we present evidence in the context of the "microglial dysregulation hypothesis", which leads to the reduction of the protective functions and increased cytotoxicity of microglia, to discuss the mechanisms involved in neurodegenerative changes and Alzheimer's disease.
Collapse
Affiliation(s)
- Juan José Triviño
- Department of Neurology, Pontificia Universidad Católica de Chile School of Medicine, Laboratory of Neuroscience. Marcoleta 391, Santiago, Chile
| | - Rommy von Bernhardi
- Department of Neurology, Pontificia Universidad Católica de Chile School of Medicine, Laboratory of Neuroscience. Marcoleta 391, Santiago, Chile; Faculty of Health Sciences, Universidad San Sebastián, Lota 2465, Santiago, Chile.
| |
Collapse
|
31
|
Virgen-Carrillo CA, de Los Ríos DLH, Torres KR, Moreno AGM. Diagnostic Criteria for Metabolic Syndrome in Diet-Induced Rodent Models: A Systematic Review. Curr Diabetes Rev 2021; 17:e140421192834. [PMID: 33855947 DOI: 10.2174/1573399817666210414103730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 01/14/2021] [Accepted: 02/09/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Thousands of publications in recent years have addressed the induction of metabolic syndrome (MetS) in rodents. However, the criteria and the reference values for diagnosing this disease have not been defined. OBJECTIVE Our main objective was to carry out a systematic review to gather evidence about the criteria for biochemical and anthropometric parameters in which scientific studies have relied on to report that rats developed MetS from a previous dietary manipulation. METHODS We compiled characteristics and findings of diet-induced MetS with high-fat, high-carbohydrate, high-fat/high-carbohydrates, and cafeteria diet from PubMed and Science Direct databases published in the last 5 years. RESULTS The results on the principal determinants for the syndrome, published in the reviewed articles, were chosen to propose reference values in the rat models of food induction. CONCLUSION The values obtained will serve as reference cut-of points in the development of the disease; in addition, the compilation of data will be useful in planning and executing research protocols in animal models.
Collapse
Affiliation(s)
- Carmen Alejandrina Virgen-Carrillo
- Instituto de Investigaciones en Comportamiento Alimentario y Nutrición, Centro Universitario del Sur, Universidad de Guadalajara, Jalisco, Mexico
| | - Diana Laura Hernández de Los Ríos
- Instituto de Investigaciones en Comportamiento Alimentario y Nutrición, Centro Universitario del Sur, Universidad de Guadalajara, Jalisco, Mexico
| | - Karina Ruíz Torres
- Instituto de Investigaciones en Comportamiento Alimentario y Nutrición, Centro Universitario del Sur, Universidad de Guadalajara, Jalisco, Mexico
| | - Alma Gabriela Martínez Moreno
- Instituto de Investigaciones en Comportamiento Alimentario y Nutrición, Centro Universitario del Sur, Universidad de Guadalajara, Jalisco, Mexico
| |
Collapse
|
32
|
Shwe T, Bo-Htay C, Leech T, Ongnok B, Jaiwongkum T, Kerdphoo S, Palee S, Pratchayasakul W, Chattipakorn N, Chattipakorn SC. D-galactose-induced aging does not cause further deterioration in brain pathologies and cognitive decline in the obese condition. Exp Gerontol 2020; 138:111001. [DOI: 10.1016/j.exger.2020.111001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 10/24/2022]
|
33
|
Manukjan N, Ahmed Z, Fulton D, Blankesteijn WM, Foulquier S. A Systematic Review of WNT Signaling in Endothelial Cell Oligodendrocyte Interactions: Potential Relevance to Cerebral Small Vessel Disease. Cells 2020; 9:cells9061545. [PMID: 32630426 PMCID: PMC7349551 DOI: 10.3390/cells9061545] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/21/2022] Open
Abstract
Key pathological features of cerebral small vessel disease (cSVD) include impairment of the blood brain barrier (BBB) and the progression of white matter lesions (WMLs) amongst other structural lesions, leading to the clinical manifestations of cSVD. The function of endothelial cells (ECs) is of major importance to maintain a proper BBB. ECs interact with several cell types to provide structural and functional support to the brain. Oligodendrocytes (OLs) myelinate axons in the central nervous system and are crucial in sustaining the integrity of white matter. The interplay between ECs and OLs and their precursor cells (OPCs) has received limited attention yet seems of relevance for the study of BBB dysfunction and white matter injury in cSVD. Emerging evidence shows a crosstalk between ECs and OPCs/OLs, mediated by signaling through the Wingless and Int-1 (WNT)/β-catenin pathway. As the latter is involved in EC function (e.g., angiogenesis) and oligodendrogenesis, we reviewed the role of WNT/β-catenin signaling for both cell types and performed a systematic search to identify studies describing a WNT-mediated interplay between ECs and OPCs/OLs. Dysregulation of this interaction may limit remyelination of WMLs and render the BBB leaky, thereby initiating a vicious neuroinflammatory cycle. A better understanding of the role of this signaling pathway in EC-OL crosstalk is essential in understanding cSVD development.
Collapse
Affiliation(s)
- Narek Manukjan
- Department of Pharmacology and Toxicology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands; or (W.M.B.)
- CARIM—School for Cardiovascular Diseases, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (Z.A.); (D.F.)
| | - Zubair Ahmed
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (Z.A.); (D.F.)
| | - Daniel Fulton
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (Z.A.); (D.F.)
| | - W. Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands; or (W.M.B.)
- CARIM—School for Cardiovascular Diseases, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands; or (W.M.B.)
- CARIM—School for Cardiovascular Diseases, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, MHeNs—School for Mental Health and Neuroscience, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
- Correspondence: ; Tel.: +31-43-3881409
| |
Collapse
|
34
|
Diaz A, Muñoz-Arenas G, Caporal-Hernandez K, Vázquez-Roque R, Lopez-Lopez G, Kozina A, Espinosa B, Flores G, Treviño S, Guevara J. Gallic acid improves recognition memory and decreases oxidative-inflammatory damage in the rat hippocampus with metabolic syndrome. Synapse 2020; 75:e22186. [PMID: 32780904 DOI: 10.1002/syn.22186] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/28/2020] [Accepted: 08/07/2020] [Indexed: 12/20/2022]
Abstract
Metabolic syndrome (MS) results from excessive consumption of high-calorie foods and sedentary lifestyles. Clinically, insulin resistance, abdominal obesity, hyperglycemia, dyslipidemia, and hypertension are observed. MS has been considered a risk factor in the development of dementia. In the brain, a metabolically impaired environment generates oxidative stress and excessive production of pro-inflammatory cytokines that deteriorate the morphology and neuronal function in the hippocampus, leading to cognitive impairment. Therapeutic alternatives suggest that phenolic compounds can be part of the treatment for neuropathies and metabolic diseases. In recent years, the use of Gallic Acid (GA) has demonstrated antioxidant and anti-inflammatory effects that contribute to neuroprotection and memory improvement in animal models. However, the effect of GA on hippocampal neurodegeneration and memory impairment under MS conditions is still unclear. In this work, we administered GA (20 mg/kg) for 60 days to rats with MS. The results show that GA treatment improved zoometric and biochemical parameters, as well as the recognition memory, in animals with MS. Additionally, GA administration increased hippocampal dendritic spines and decreased oxidative stress and inflammation. Our results show that GA treatment improves metabolism: reducing the oxidative and inflammatory environment that facilitates the recovery of the neuronal morphology in the hippocampus of rats with MS. Consequently, the recognition of objects by these animals, suggesting that GA could be used therapeutically in metabolic disorders that cause dementia.
Collapse
Affiliation(s)
- Alfonso Diaz
- Facultad de Ciencias Quimicas, Benemerita Universidad Autonoma de Puebla, Puebla, Mexico
| | - Guadalupe Muñoz-Arenas
- Facultad de Ciencias Quimicas, Benemerita Universidad Autonoma de Puebla, Puebla, Mexico
| | | | - Rubén Vázquez-Roque
- Laboratorio de Neuropsiquiatria, Instituto de Fisiologia, Benemerita Universidad Autonoma de Puebla, Puebla, Mexico
| | - Gustavo Lopez-Lopez
- Facultad de Ciencias Quimicas, Benemerita Universidad Autonoma de Puebla, Puebla, Mexico
| | - Anna Kozina
- Instituto de Química, Universidad Nacional Autonoma de México, Ciudad de Mexico, Mexico
| | - Blanca Espinosa
- Departamento de Bioquimica, Instituto Nacional de Enfermedades Respiratorias, ICV, Ciudad de Mexico, Mexico
| | - Gonzalo Flores
- Laboratorio de Neuropsiquiatria, Instituto de Fisiologia, Benemerita Universidad Autonoma de Puebla, Puebla, Mexico
| | - Samuel Treviño
- Facultad de Ciencias Quimicas, Benemerita Universidad Autonoma de Puebla, Puebla, Mexico
| | - Jorge Guevara
- Departamento de Bioquimica, Facultad de Medicina, Universidad Nacional Autonoma de México, Ciudad de Mexico, Mexico
| |
Collapse
|