1
|
Elzinga SE, Guo K, Turfah A, Henn RE, Webber‐Davis IF, Hayes JM, Pacut CM, Teener SJ, Carter AD, Rigan DM, Allouch AM, Jang D, Parent R, Glass E, Murphy GG, Lentz SI, Chen KS, Zhao L, Hur J, Feldman EL. Metabolic stress and age drive inflammation and cognitive decline in mice and humans. Alzheimers Dement 2025; 21:e70060. [PMID: 40110679 PMCID: PMC11923576 DOI: 10.1002/alz.70060] [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: 09/19/2024] [Revised: 01/17/2025] [Accepted: 01/31/2025] [Indexed: 03/22/2025]
Abstract
INTRODUCTION Metabolic stressors (obesity, metabolic syndrome, prediabetes, and type 2 diabetes [T2D]) increase the risk of cognitive impairment (CI), including Alzheimer's disease (AD). Immune system dysregulation and inflammation, particularly microglial mediated, may underlie this risk, but mechanisms remain unclear. METHODS Using a high-fat diet-fed (HFD) model, we assessed longitudinal metabolism and cognition, and terminal inflammation and brain spatial transcriptomics. Additionally, we performed hippocampal spatial transcriptomics and single-cell RNA sequencing of post mortem tissue from AD and T2D human subjects versus controls. RESULTS HFD induced progressive metabolic and CI with terminal inflammatory changes, and dysmetabolic, neurodegenerative, and inflammatory gene expression profiles, particularly in microglia. AD and T2D human subjects had similar gene expression changes, including in secreted phosphoprotein 1 (SPP1), a pro-inflammatory gene associated with AD. DISCUSSION These data show that metabolic stressors cause early and progressive CI, with inflammatory changes that promote disease. They also indicate a role for microglia, particularly microglial SPP1, in CI. HIGHLIGHTS Metabolic stress causes persistent metabolic and cognitive impairments in mice. Murine and human brain spatial transcriptomics align and indicate a pro-inflammatory milieu. Transcriptomic data indicate a role for microglial-mediated inflammatory mechanisms. Secreted phosphoprotein 1 emerged as a potential target of interest in metabolically driven cognitive impairment.
Collapse
Affiliation(s)
- Sarah E. Elzinga
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
- Department of PhysiologyMichigan State UniversityEast LansingMichiganUSA
| | - Kai Guo
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Ali Turfah
- Department of BiostatisticsSchool of Public HealthUniversity of MichiganAnn ArborMichiganUSA
| | - Rosemary E. Henn
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | | | - John M. Hayes
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Crystal M. Pacut
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Samuel J. Teener
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Andrew D. Carter
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Diana M. Rigan
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Adam M. Allouch
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Dae‐Gyu Jang
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Rachel Parent
- Department of Internal MedicineGeneral MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Emily Glass
- Department of Molecular and Integrative PhysiologyDivision of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Geoffrey G. Murphy
- Department of Molecular and Integrative PhysiologyDivision of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Stephen I. Lentz
- Department of Internal MedicineDivision of MetabolismEndocrinology, and DiabetesUniversity of MichiganAnn ArborMichiganUSA
| | - Kevin S. Chen
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
- Department of NeurosurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Lili Zhao
- Department of BiostatisticsSchool of Public HealthUniversity of MichiganAnn ArborMichiganUSA
| | - Junguk Hur
- Department of Biomedical SciencesUniversity of North DakotaGrand ForksNorth DakotaUSA
| | - Eva L. Feldman
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
2
|
Nazeam JA, Black I, Mulamoottil VA, Selim NM, El Shiekh RA, Abu-Elfotuh K, Hamdan AME, Gowifel AMH, Hafez SM, Mohamed EK, Atwa AM, El Hefnawy HM, Azadi P. Okra seed polysaccharides mitigate neuroinflammation and cognitive impairment via modulation of Nrf2/HO-1, HMGB1/RAGE/TLR4/NF-κB, NLRP3/Caspase-1, JAK-2/STAT-3, AMPK/SIRT1/m-TOR, PI3K/AKT/CREB/BDNF/TrkB and PERK/CHOP/Bcl-2 axes. Int Immunopharmacol 2025; 148:114110. [PMID: 39862637 DOI: 10.1016/j.intimp.2025.114110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/23/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
Global healthcare systems are under tremendous strain due to the increasing prevalence of neurodegenerative disorders. Growing data suggested that overconsumption of high-fat/high-carbohydrates diet (HFHCD) is associated with enhanced incidence of metabolic alterations, neurodegeneration, and cognitive dysfunction. Functional foods have gained prominence in curbing metabolic and neurological deficits. Consequently, this study endeavored to explore effects of purified Okra seed polysaccharides (OP) (Abelmoschus esculentus (L.) Moench) against HFHCD-induced metabolic alterations and cognitive dysfunction, with elucidating underlying contributed mechanistic pathways. OP hydrolysate was analyzed using GC-MS analysis. The biological study encompassed two phases, the first phase I (model establishment phase), for 3 months, involved a control group, fed standard diet, and HFHCD group. The second phase (phase II) where HFHCD fed rats were re-divided into 3 equal subgroups, 1st subgroup received HFHCD, whereas second and third subgroups received OP, 200 or 400 mg/kg/day, respectively, for 28 days. GC-MS characterized OP as an arabinogalactouranan and revealed the monosaccharide composition as galacturonic acid: arabinose: glucose: galactose: rhamnose: xylose in ratio of 28.2: 23.3: 11.5: 4.2: 3.5: 2.0. The findings demonstrated that OP dose-dependently mitigated HFHCD-induced rise in body weights, lipid profiles, levels of blood glucose and disruption in behavioral outcomes, neurotransmitters, together with histopathological alterations in brain. Moreover, OP dose-dependently improved redox, neuroinflammatory, endoplasmic reticulum (ER) stress, autophagic and apoptotic biomarkers. OP can be regarded as promising functional food candidate to hamper HFHCD-induced metabolic alterations and cognitive deficit, via enhancing Nrf2/HO-1, AMPK/SIRT1 and PI3K/AKT/CREB axes, long with dampening of HMGB1/RAGE/TLR4, NLRP3/Caspase-1, JAK-2/STAT-3 and PERK/CHOP axes.
Collapse
Affiliation(s)
- Jilan A Nazeam
- Department of Pharmacognosy, Faculty of Pharmacy, October 6 University, Giza, Egypt; Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA.
| | - Ian Black
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA.
| | | | - Nabil M Selim
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Giza 11562, Egypt.
| | - Riham A El Shiekh
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Giza 11562, Egypt.
| | - Karema Abu-Elfotuh
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt; Al-Ayen Iraqi University, Thi-Qar 64001, Iraq.
| | - Ahmed M E Hamdan
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; Prince Fahad bin Sultan Chair for Biomedical Research, University of Tabuk, Saudi Arabia.
| | - Ayah M H Gowifel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo 11571, Egypt.
| | - Shaimaa M Hafez
- Department of Anatomy and Embryology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt.
| | - Ehsan K Mohamed
- Biochemistry Department, Egyptian Drug Authority (EDA), Formerly National Organization of Drug Control and Research (NODCAR), Giza, Egypt
| | - Ahmed M Atwa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo-Suez Road, Cairo 11829, Egypt; Department of Pharmacology and Toxicology, College of Pharmacy, Al-Ayen Iraqi University, Thi-Qar, 64001, Iraq.
| | | | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA.
| |
Collapse
|
3
|
Jo D, Choi SY, Ahn SY, Song J. IGF1 enhances memory function in obese mice and stabilizes the neural structure under insulin resistance via AKT-GSK3β-BDNF signaling. Biomed Pharmacother 2025; 183:117846. [PMID: 39805192 DOI: 10.1016/j.biopha.2025.117846] [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: 09/25/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/16/2025] Open
Abstract
Obesity is a prevalent metabolic disorder linked to insulin resistance, hyperglycemia, increased adiposity, chronic inflammation, and cognitive dysfunction. Recent research has focused on developing therapeutic strategies to mitigate cognitive impairment associated with obesity. Insulin growth factor-1 (IGF1) deficiency is linked to insulin resistance, glucose intolerance, and the progression of obesity-related central nervous system (CNS) disorders. In this study, we investigated the neuroprotective effects of IGF1 in two obesity models: diet-induced obesity (high-fat diet mice) and genetic obesity (ob/ob mice which is genetically deficient in leptin), and in vitro Neuro2A neuronal cells and primary cortical neurons under insulin resistance conditions. We performed RNA sequencing analysis using the cortex of high-fat diet mice injected with IGF1. Also, we detected cytokine levels in blood of high-fat diet mice injected with IGF1. In addition, we conducted the Barnes maze test as a spatial memory function test and open field test as an anxiety behavior test in ob/ob mice. We measured the levels of proteins and mRNAs related to insulin signaling, including synaptic density proteins in brain cortex of ob/ob mice. Our results showed that IGF1 injection enhanced spatial memory function and synaptic plasticity in obese mice. Furthermore, in vitro data demonstrated that IGF1 treated neurons revealed enhanced neural complexity and improved neurite outgrowth under insulin resistance condition through the AKT-GSK3β-BDNF pathway related to antidepressant, cognitive function and anti-apoptotic mechanisms. Therefore, our results provided that IGF1 have potential to alleviate cognitive impairment by promoting synaptic plasticity and neural complexity in the obese brain.
Collapse
Affiliation(s)
- Danbi Jo
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Republic of Korea.
| | - Seo Yoon Choi
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Republic of Korea; Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Republic of Korea.
| | - Seo Yeon Ahn
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Republic of Korea; Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Republic of Korea.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Republic of Korea; Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Republic of Korea.
| |
Collapse
|
4
|
Cao X, Wang Q, Zhang L, Sun H, Xu G, Chen X, Wu Z, Liu H, Yuan G, Wu J, Liu T. Oxytocin alleviates high-fat diet-induced anxiety by decreasing glutamatergic synaptic transmission in the ventral dentate gyrus in adolescent mice. Neuropharmacology 2025; 262:110201. [PMID: 39481751 DOI: 10.1016/j.neuropharm.2024.110201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 09/21/2024] [Accepted: 10/27/2024] [Indexed: 11/02/2024]
Abstract
A high-fat diet (HFD)-induced obesity is associated with mental disorders in adolescence. However, the mechanisms underlying these associations remain unclear. In this study, we hypothesized that synaptic remodeling occurs in the ventral hippocampus (vHP) of obese mice. To investigate this, we established a postnatal model of HFD-induced obesity in mice and observed increased body weight, elevated plasma luteinizing hormone and testosterone levels, premature puberty, and enhanced anxiety-like behavior in male subjects. We also examined the effect of HFD on the c-Fos protein expression in the ventral dentate gyrus (vDG) and explored the influence of intracerebroventricular (i.c.v) oxytocin injections on HFD-induced anxiety. Our results indicated an increase in c-Fos-positive cells in the vDG following HFD consumption. Additionally, we recorded the spontaneous synaptic activity of miniature excitatory postsynaptic currents (mEPSCs) in the vDG. Notably, HFD resulted in an elevated mEPSC frequency without affecting mEPSC amplitude. Subsequently, investigations demonstrated that i.c.v oxytocin injections reversed anxiety-like behavior induced by HFD. Moreover, the application of oxytocin in a bath solution reduced the mEPSC frequency in the vDG. These findings suggest that postnatal HFD intake induces synaptic dysfunction in the vDG, associated with the hyperactivity of vDG neurons, potentially contributing to the anxiety-like behavior in juvenile obesity.
Collapse
Affiliation(s)
- Xi Cao
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Qiyuan Wang
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Lina Zhang
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Huichao Sun
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Gang Xu
- Jiangxi Provincial Key Laboratory of Trauma, Burn and Pain Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Xiao Chen
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Zhihong Wu
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Huibao Liu
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Gaole Yuan
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| | - Jian Wu
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| | - Tao Liu
- Department of Pediatrics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China; Jiangxi Provincial Key Laboratory of Trauma, Burn and Pain Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
5
|
Machado AE, Rezer P, Mancini G, Latini A, Moreira ELG. Short-term high-fat diet alters behavior, peripheral metabolism, and brain mitochondrial function in Swiss mice. AN ACAD BRAS CIENC 2024; 96:e20240880. [PMID: 39661818 DOI: 10.1590/0001-3765202420240880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/03/2024] [Indexed: 12/13/2024] Open
Abstract
Understanding the temporal dynamics of high-fat diets (HFD) effects on behavior and metabolism is crucial for comprehending their negative impact on organisms. This study investigated the short-term effects (15, 25, and 35 days) of HFD in Swiss mice. Our findings revealed distinct behavioral and metabolic changes throughout the treatment. After 15 days of HFD, mice exhibited impaired exploratory habituation and significant increases in visceral adipose mass, fasting glucose levels, and glucose intolerance. Extending the diet to 25 days intensified the metabolic effects, resulting in compromised acquisition of recognition memory, increased body mass gain, and elevated plasma total cholesterol and triglyceride levels. After 35 days of HFD, these effects were further intensified and accompanied by anxiogenic-like responses in the open field test. Additionally, we observed a positive correlation between metabolic changes and behavioral impairments alongside prefrontal cortex mitochondrial dysfunction. In conclusion, our study reveals the temporal dynamics of behavioral and metabolic changes induced by short-term HFD in Swiss mice, highlighting the relationship between metabolic dysfunction and behavioral impairments. These findings pave the way for future research to unravel the underlying mechanisms and develop strategies to counteract the detrimental effects of HFD on behavior and metabolism.
Collapse
Affiliation(s)
- Adriano Emanuel Machado
- Universidade Federal de Santa Catarina, Programa de Pós-Graduação em Neurociências, Campus Universitário, s/n, Córrego Grande, 88040-900 Florianópolis, SC, Brazil
| | - Peterson Rezer
- Universidade Federal de Santa Catarina, Programa de Pós-Graduação em Neurociências, Campus Universitário, s/n, Córrego Grande, 88040-900 Florianópolis, SC, Brazil
| | - Gianni Mancini
- Universidade Federal de Santa Catarina, Programa de Pós-Graduação em Neurociências, Campus Universitário, s/n, Córrego Grande, 88040-900 Florianópolis, SC, Brazil
| | - Alexandra Latini
- Universidade Federal de Santa Catarina, Centro de Ciências Biológicas, Departamento de Bioquímica, Laboratório de Bioenergética e Estresse Oxidativo - LABOX, Campus Universitário - Trindade, Rua da Prefeitura Universitária, s/n, Bairro Córrego Grande, 88037-000 Florianópolis, SC, Brazil
| | - Eduardo Luiz G Moreira
- Universidade Federal de Santa Catarina, Centro de Ciências Biológicas, Departamento de Ciências Fisiológicas, Campus Universitário, Rua da Prefeitura Universitária, s/n, Córrego Grande, 88037-000 Florianópolis, SC, Brazil
- Universidade Federal de Santa Catarina, Programa de Pós-Graduação em Neurociências, Campus Universitário, s/n, Córrego Grande, 88040-900 Florianópolis, SC, Brazil
| |
Collapse
|
6
|
Shaabani Ghahremanlo M, Hojati V, Vaezi G, Sharafi S. Transgenerational of Oxidative Damage Induced by Prenatal Ethanol Exposure on Spatial Learning/Memory and BDNF in the of Male Rats. IBRO Neurosci Rep 2024; 17:398-406. [PMID: 39583589 PMCID: PMC11583777 DOI: 10.1016/j.ibneur.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 09/11/2024] [Indexed: 11/26/2024] Open
Abstract
Alcohol consumption during pregnancy harms fetal development, leading to various physical and behavioral issues. This study investigates how prenatal ethanol exposure triggers oxidative stress (OS) and affects neurotrophic factors (NTFs), particularly brain-derived growth factor (BDNF) gene expression in the hippocampus, influencing learning and memory decline across two generations of male offspring from ethanol-exposed female rats. A rat model of fetal alcohol spectrum disorder (FASD) was initially generated to reflect on the deficits in the first generation, and then those transmitted via the male germline to the unexposed male ones. The pregnant rats were thus divided into four groups, namely, the control group (CTRL) receiving only distilled water (DW), and three groups being exposed to ethanol (20 %, 4.5 g/kg) by oral gavage, during the first 10-day gestation (FG), the second 10-day gestation (SG), and the entire gestation (EG) periods. Subsequent Morris water maze (MWM) tests on male offspring revealed spatial learning deficits during the second and entire gestational periods in both generations. Analysis of antioxidant enzyme activity including glutathione peroxidase (GPx), superoxide dismutase (SOD), and malondialdehyde (MDA), and BDNF gene expression in the hippocampus further highlighted the impacts of prenatal ethanol exposure. The study results demonstrated that prenatal ethanol exposure caused spatial learning/memory deficits during the SG and EG, altered antioxidant enzyme activity, and reduced BDNF gene expression in both generations. The findings underscore the role of OS in developmental and behavioral issues in FASD rat models and suggest that lasting transgenerational effects in the second generation may stem from alcohol-induced changes.
Collapse
Affiliation(s)
| | | | - Gholamhassan Vaezi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Shahram Sharafi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| |
Collapse
|
7
|
N'Diaye M, Ducourneau EG, Bakoyiannis I, Potier M, Lafenetre P, Ferreira G. Obesogenic diet induces sex-specific alterations of contextual fear memory and associated hippocampal activity in mice. Cereb Cortex 2024; 34:bhae254. [PMID: 38934712 DOI: 10.1093/cercor/bhae254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
In addition to metabolic and cardiovascular disorders, obesity is associated with cognitive deficits in humans and animal models. We have previously shown that obesogenic high-fat and sugar diet intake during adolescence (adoHFSD) impairs hippocampus (HPC)-dependent memory in rodents. These results were obtained in males only and it remains to evaluate whether adoHFSD has similar effect in females. Therefore, here, we investigated the effects of adoHFSD consumption on HPC-dependent contextual fear memory and associated brain activation in male and female mice. Exposure to adoHFSD increased fat mass accumulation and glucose levels in both males and females but impaired contextual fear memory only in males. Compared with females, contextual fear conditioning induced higher neuronal activation in the dorsal and ventral HPC (CA1 and CA3 subfields) as well as in the medial prefrontal cortex in males. Also, adoHFSD-fed males showed enhanced c-Fos expression in the dorsal HPC, particularly in the dentate gyrus, and in the basolateral amygdala compared with the other groups. Finally, chemogenetic inactivation of the dorsal HPC rescued adoHFSD-induced memory deficits in males. Our results suggest that males are more vulnerable to the effects of adoHFSD on HPC-dependent aversive memory than females, due to overactivation of the dorsal HPC.
Collapse
Affiliation(s)
- Matéo N'Diaye
- NutriNeuro Lab, FoodCircus Team, Université de Bordeaux, UMR 1286 INRAE, Bordeaux INP, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Eva-Gunnel Ducourneau
- NutriNeuro Lab, FoodCircus Team, Université de Bordeaux, UMR 1286 INRAE, Bordeaux INP, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Ioannis Bakoyiannis
- NutriNeuro Lab, FoodCircus Team, Université de Bordeaux, UMR 1286 INRAE, Bordeaux INP, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Mylène Potier
- NutriNeuro Lab, FoodCircus Team, Université de Bordeaux, UMR 1286 INRAE, Bordeaux INP, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Pauline Lafenetre
- NutriNeuro Lab, FoodCircus Team, Université de Bordeaux, UMR 1286 INRAE, Bordeaux INP, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Guillaume Ferreira
- NutriNeuro Lab, FoodCircus Team, Université de Bordeaux, UMR 1286 INRAE, Bordeaux INP, 146 rue Léo Saignat, 33076 Bordeaux, France
| |
Collapse
|
8
|
Anderson T, Sharma S, Kelberman MA, Ware C, Guo N, Qin Z, Weinshenker D, Parent MB. Obesity during preclinical Alzheimer's disease development exacerbates brain metabolic decline. J Neurochem 2024; 168:801-821. [PMID: 37391269 DOI: 10.1111/jnc.15900] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/29/2023] [Accepted: 06/13/2023] [Indexed: 07/02/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Obesity in middle age increases AD risk and severity, which is alarming given that obesity prevalence peaks at middle age and obesity rates are accelerating worldwide. Midlife, but not late-life obesity increases AD risk, suggesting that this interaction is specific to preclinical AD. AD pathology begins in middle age, with accumulation of amyloid beta (Aβ), hyperphosphorylated tau, metabolic decline, and neuroinflammation occurring decades before cognitive symptoms appear. We used a transcriptomic discovery approach in young adult (6.5 months old) male and female TgF344-AD rats that overexpress mutant human amyloid precursor protein and presenilin-1 and wild-type (WT) controls to determine whether inducing obesity with a high-fat/high-sugar "Western" diet during preclinical AD increases brain metabolic dysfunction in dorsal hippocampus (dHC), a brain region vulnerable to the effects of obesity and early AD. Analyses of dHC gene expression data showed dysregulated mitochondrial and neurotransmission pathways, and up-regulated genes involved in cholesterol synthesis. Western diet amplified the number of genes that were different between AD and WT rats and added pathways involved in noradrenergic signaling, dysregulated inhibition of cholesterol synthesis, and decreased intracellular lipid transporters. Importantly, the Western diet impaired dHC-dependent spatial working memory in AD but not WT rats, confirming that the dietary intervention accelerated cognitive decline. To examine later consequences of early transcriptional dysregulation, we measured dHC monoamine levels in older (13 months old) AD and WT rats of both sexes after long-term chow or Western diet consumption. Norepinephrine (NE) abundance was significantly decreased in AD rats, NE turnover was increased, and the Western diet attenuated the AD-induced increases in turnover. Collectively, these findings indicate obesity during prodromal AD impairs memory, potentiates AD-induced metabolic decline likely leading to an overproduction of cholesterol, and interferes with compensatory increases in NE transmission.
Collapse
Affiliation(s)
- Thea Anderson
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Sumeet Sharma
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael A Kelberman
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Christopher Ware
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Nanxi Guo
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Georgia, USA
| | - Zhaohui Qin
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Georgia, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Marise B Parent
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
- Department of Psychology, Georgia State University, Georgia, USA
| |
Collapse
|
9
|
Hernandez Torres LD, Rezende F, Peschke E, Will O, Hövener JB, Spiecker F, Özorhan Ü, Lampe J, Stölting I, Aherrahrou Z, Künne C, Kusche-Vihrog K, Matschl U, Hille S, Brandes RP, Schwaninger M, Müller OJ, Raasch W. Incidence of microvascular dysfunction is increased in hyperlipidemic mice, reducing cerebral blood flow and impairing remote memory. Front Endocrinol (Lausanne) 2024; 15:1338458. [PMID: 38469142 PMCID: PMC10925718 DOI: 10.3389/fendo.2024.1338458] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/24/2024] [Indexed: 03/13/2024] Open
Abstract
Introduction The development of cognitive dysfunction is not necessarily associated with diet-induced obesity. We hypothesized that cognitive dysfunction might require additional vascular damage, for example, in atherosclerotic mice. Methods We induced atherosclerosis in male C57BL/6N mice by injecting AAV-PCSK9DY (2x1011 VG) and feeding them a cholesterol-rich Western diet. After 3 months, mice were examined for cognition using Barnes maze procedure and for cerebral blood flow. Cerebral vascular morphology was examined by immunehistology. Results In AAV-PCSK9DY-treated mice, plaque burden, plasma cholesterol, and triglycerides are elevated. RNAseq analyses followed by KEGG annotation show increased expression of genes linked to inflammatory processes in the aortas of these mice. In AAV-PCSK9DY-treated mice learning was delayed and long-term memory impaired. Blood flow was reduced in the cingulate cortex (-17%), caudate putamen (-15%), and hippocampus (-10%). Immunohistological studies also show an increased incidence of string vessels and pericytes (CD31/Col IV staining) in the hippocampus accompanied by patchy blood-brain barrier leaks (IgG staining) and increased macrophage infiltrations (CD68 staining). Discussion We conclude that the hyperlipidemic PCSK9DY mouse model can serve as an appropriate approach to induce microvascular dysfunction that leads to reduced blood flow in the hippocampus, which could explain the cognitive dysfunction in these mice.
Collapse
Affiliation(s)
| | - Flavia Rezende
- Institute for Cardiovascular Physiology, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
- DZHK (German Center for Cardiovascular Research) Partner Site Rhine-Main, Germany
| | - Eva Peschke
- Section Biomedical Imaging, Molecular Imaging North Competence Center (MOIN CC), Department of Radiology and Neuroradiology, Universitätsklinikum Schleswig-Holstein (UKSH), Kiel University, Kiel, Germany
| | - Olga Will
- Section Biomedical Imaging, Molecular Imaging North Competence Center (MOIN CC), Department of Radiology and Neuroradiology, Universitätsklinikum Schleswig-Holstein (UKSH), Kiel University, Kiel, Germany
| | - Jan-Bernd Hövener
- Section Biomedical Imaging, Molecular Imaging North Competence Center (MOIN CC), Department of Radiology and Neuroradiology, Universitätsklinikum Schleswig-Holstein (UKSH), Kiel University, Kiel, Germany
| | - Frauke Spiecker
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Ümit Özorhan
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Josephine Lampe
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Ines Stölting
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Zouhair Aherrahrou
- Institute for Cardiogenetics, University Lübeck; University of Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany
| | - Carsten Künne
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Kristina Kusche-Vihrog
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany
- Institute for Physiology, University Lübeck, Lübeck, Germany
| | - Urte Matschl
- Department Virus Immunology, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Susanne Hille
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany
- Department of Internal Medicine III, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Ralf P. Brandes
- Institute for Cardiovascular Physiology, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
- DZHK (German Center for Cardiovascular Research) Partner Site Rhine-Main, Germany
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany
- CBBM (Centre for Brain, Behavior and Metabolism), University of Lübeck, Lübeck, Germany
| | - Oliver J. Müller
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany
- Department of Internal Medicine III, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Walter Raasch
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany
- CBBM (Centre for Brain, Behavior and Metabolism), University of Lübeck, Lübeck, Germany
| |
Collapse
|
10
|
Herrera G, Silvero C MJ, Becerra MC, Lasaga M, Scimonelli T. Modulatory role of α-MSH in hippocampal-dependent memory impairment, synaptic plasticity changes, oxidative stress, and astrocyte reactivity induced by short-term high-fat diet intake. Neuropharmacology 2023; 239:109688. [PMID: 37591460 DOI: 10.1016/j.neuropharm.2023.109688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/11/2023] [Accepted: 08/12/2023] [Indexed: 08/19/2023]
Abstract
High-fat diet (HFD) consumption is associated with cognitive deficits and neurodegenerative diseases. Since the hippocampus is extremely sensitive to pathophysiological changes, neuroinflammation and the concomitant oxidative stress induced by HFD can significantly interfere with hippocampal-dependent functions related to learning and memory. The neuropeptide alpha-melanocyte stimulating hormone (α-MSH) mediates neuroprotective actions in the central nervous system and can reverse the effects of neuroinflammation in cognitive functions that depend on the hippocampus. In this study, we used male Wistar rats to evaluate the effect of short-term HFD intake (5 days) plus a mild immune challenge, Lipopolysaccharide (LPS 10 μg/kg) on contextual fear, changes in structural plasticity, oxidative stress, and astrocyte reactivation in the hippocampus. We also determined the possible modulatory role of α-MSH. HFD consumption was associated with an increase in markers of oxidative stress (Advanced oxidation protein products and Malondialdehyde) in the dorsal hippocampus (DH). We also found changes in hippocampal structural synaptic plasticity, observing a decrease in total spine in the DH after HFD plus LPS. We observed astrocyte proliferation and a significant increase in the percentage of the area occupied by GFAP. Treatment with α-MSH (0.1 μg/0.25 μl) in the DH reversed the effect of short-term HFD plus LPS on contextual fear memory, oxidative stress, and spine density. α-MSH also reduced astrocyte proliferation. Our present results indicate that HFD consumption for a short period sensitizes the central nervous system (CNS) to a subsequent immune challenge and impairs contextual fear memory and that α-MSH could have a modulatory protective effect.
Collapse
Affiliation(s)
- Guadalupe Herrera
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET. Departamento de Farmacología Otto Orshinger, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.
| | - M Jazmín Silvero C
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica UNITEFA-CONICET. Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - M Cecilia Becerra
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica UNITEFA-CONICET. Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mercedes Lasaga
- Instituto de Investigaciones Biomédicas INBIOMED UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Teresa Scimonelli
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET. Departamento de Farmacología Otto Orshinger, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
11
|
da Silva LE, de Oliveira MP, da Silva MR, Abel JDS, Tartari G, de Aguiar da Costa M, Ludvig Gonçalves C, Rezin GT. L-carnitine and Acetyl-L Carnitine: A Possibility for Treating Alterations Induced by Obesity in the Central Nervous System. Neurochem Res 2023; 48:3316-3326. [PMID: 37495838 DOI: 10.1007/s11064-023-04000-z] [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: 03/04/2023] [Revised: 06/21/2023] [Accepted: 07/21/2023] [Indexed: 07/28/2023]
Abstract
Excessive consumption of nutrients, as well as obesity, leads to an inflammatory process, especially in adipose tissue. This inflammation reaches the systemic level and, subsequently, the central nervous system (CNS), which can lead to oxidative stress and mitochondrial dysfunction, resulting in brain damage. Thus, adequate treatment for obesity is necessary, including lifestyle changes (diet adequation and physical activity) and pharmacotherapy. However, these drugs can adversely affect the individual's health. In this sense, searching for new therapeutic alternatives for reestablishing metabolic homeostasis is necessary. L-carnitine (LC) and acetyl-L-carnitine (LAC) have neuroprotective effects against oxidative stress and mitochondrial dysfunction in several conditions, including obesity. Therefore, this study aimed to conduct a narrative review of the literature on the effect of LC and LAC on brain damage caused by obesity, in particular, on mitochondrial dysfunction and oxidative stress. Overall, these findings highlight that LC and LAC may be a promising treatment for recovering REDOX status and mitochondrial dysfunction in the CNS in obesity. Future work should focus on better elucidating the molecular mechanisms behind this treatment.
Collapse
Affiliation(s)
- Larissa Espindola da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil.
| | - Mariana Pacheco de Oliveira
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Mariella Reinol da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Jéssica da Silva Abel
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Gisele Tartari
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Maiara de Aguiar da Costa
- Laboratory of Neurology, Graduate Program in Health Sciences, University of Extreme South Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Cinara Ludvig Gonçalves
- Laboratory of Neurology, Graduate Program in Health Sciences, University of Extreme South Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| |
Collapse
|
12
|
Maneechote C, Pintana H, Kerdphoo S, Janjek S, Chattipakorn N, Chattipakorn SC. Differential temporal therapies with pharmacologically targeted mitochondrial fission/fusion protect the brain against acute myocardial ischemia-reperfusion injury in prediabetic rats: The crosstalk between mitochondrial apoptosis and inflammation. Eur J Pharmacol 2023; 956:175939. [PMID: 37536625 DOI: 10.1016/j.ejphar.2023.175939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/08/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
An imbalance of brain mitochondrial dynamics, increases in brain inflammation and apoptosis, and increasing cognitive dysfunction, have been reported as being associated with prediabetes and myocardial ischemia-reperfusion (IR) injury. Since inhibiting mitochondrial fission with Mdivi-1 or promoting fusion with M1 had cardioprotective effects in myocardial IR injury and obesity, the neuroprotective roles of Mdivi-1 and M1 when administered at different time points of myocardial IR injury in obese prediabetes have never been determined. Ninety-six male Wistar rats were fed with either a normal (ND: n = 8) or a high-fat diet to induce prediabetes (HFD: n = 88) for 12 weeks. At week 13, all rats were subjected to left anterior descending coronary artery ligation for 30 min, followed by reperfusion for 120 min. HFD rats were randomly divided into 10 groups and assigned into either a pre-ischemic group treated with vehicle (HFV), pre-ischemic, during-ischemic, or onset of reperfusion groups treated with either Mdivi-1 (MDV), M1, or combined (COM). Heart function was examined invasively, with the heart being terminated to investigate myocardial infarction. Brains were collected to determine mitochondrial functions, inflammation, apoptosis, and pathological markers. Mdivi-1, M1, and COM treatment at different periods exerted cardioprotection against myocardial IR injury in HFD-fed rats by reducing infarct size and left ventricular dysfunction. All interventions also improved all brain pathologies against myocardial IR injury in prediabetic rats. These findings suggest that differential temporal modulation of mitochondrial dynamics may be appropriate regimens for preventing heart and brain complications after myocardial IR injury in obese prediabetes.
Collapse
Affiliation(s)
- Chayodom Maneechote
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Hiranya Pintana
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sasiwan Kerdphoo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sornram Janjek
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
13
|
Batabyal RA, Bansal A, Cechinel LR, Authelet K, Goldberg M, Nadler E, Keene CD, Jayadev S, Domoto-Reilly K, Li G, Peskind E, Hashimoto-Torii K, Buchwald D, Freishtat RJ. Adipocyte-Derived Small Extracellular Vesicles from Patients with Alzheimer Disease Carry miRNAs Predicted to Target the CREB Signaling Pathway in Neurons. Int J Mol Sci 2023; 24:14024. [PMID: 37762325 PMCID: PMC10530811 DOI: 10.3390/ijms241814024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Alzheimer disease (AD) is characterized by amyloid-β (Aβ) plaques, neurofibrillary tangles, synaptic dysfunction, and progressive dementia. Midlife obesity increases the risk of developing AD. Adipocyte-derived small extracellular vesicles (ad-sEVs) have been implicated as a mechanism in several obesity-related diseases. We hypothesized that ad-sEVs from patients with AD would contain miRNAs predicted to downregulate pathways involved in synaptic plasticity and memory formation. We isolated ad-sEVs from the serum and cerebrospinal fluid (CSF) of patients with AD and controls and compared miRNA expression profiles. We performed weighted gene co-expression network analysis (WGCNA) on differentially expressed miRNAs to identify highly interconnected clusters correlating with clinical traits. The WGCNA identified a module of differentially expressed miRNAs, in both the serum and CSF, that was inversely correlated with the Mini-Mental State Examination scores. Within this module, miRNAs that downregulate CREB signaling in neurons were highly represented. These results demonstrate that miRNAs carried by ad-sEVs in patients with AD may downregulate CREB signaling and provide a potential mechanistic link between midlife obesity and increased risk of AD.
Collapse
Affiliation(s)
- Rachael A. Batabyal
- Center for Genetic Medicine, Children’s National Hospital, Washington, DC 20012, USA (M.G.); (R.J.F.)
- School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA; (E.N.)
| | - Ankush Bansal
- Center for Neuroscience Research, Children’s National Hospital, Washington, DC 20010, USA
| | - Laura Reck Cechinel
- Center for Genetic Medicine, Children’s National Hospital, Washington, DC 20012, USA (M.G.); (R.J.F.)
| | - Kayla Authelet
- Center for Genetic Medicine, Children’s National Hospital, Washington, DC 20012, USA (M.G.); (R.J.F.)
| | - Madeleine Goldberg
- Center for Genetic Medicine, Children’s National Hospital, Washington, DC 20012, USA (M.G.); (R.J.F.)
| | - Evan Nadler
- School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA; (E.N.)
- Division of Pediatric Surgery, Children’s National Hospital, Washington, DC 20010, USA
| | - C. Dirk Keene
- Department of Pathology, University of Washington, Seattle, WA 98104, USA;
| | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle, WA 98104, USA; (S.J.)
| | - Kimiko Domoto-Reilly
- Department of Neurology, University of Washington, Seattle, WA 98104, USA; (S.J.)
| | - Gail Li
- Department of Psychology and Behavioral Sciences, School of Medicine, University of Washington, Seattle, WA 98104, USA
- Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Elaine Peskind
- Department of Psychology and Behavioral Sciences, School of Medicine, University of Washington, Seattle, WA 98104, USA
- Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Kazue Hashimoto-Torii
- School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA; (E.N.)
- Center for Neuroscience Research, Children’s National Hospital, Washington, DC 20010, USA
| | - Dedra Buchwald
- Institute for Research Education to Advance Community Health, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
| | - Robert J. Freishtat
- Center for Genetic Medicine, Children’s National Hospital, Washington, DC 20012, USA (M.G.); (R.J.F.)
- School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA; (E.N.)
| |
Collapse
|
14
|
Kang J, Park M, Oh CM, Kim T. High-fat diet-induced dopaminergic dysregulation induces REM sleep fragmentation and ADHD-like behaviors. Psychiatry Res 2023; 327:115412. [PMID: 37607442 DOI: 10.1016/j.psychres.2023.115412] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 07/27/2023] [Accepted: 08/10/2023] [Indexed: 08/24/2023]
Abstract
Consumption of a high-fat diet (HFD) has been associated with reduced wakefulness and various behavioral deficits, including anxiety, depression, and anhedonia. The dopaminergic system, which plays a crucial role in sleep and ADHD, is known to be vulnerable to chronic HFD. However, the association between HFD-induced behavioral and molecular changes remains unclear. Therefore, we investigated the effects of a HFD on the dopaminergic system and its association with behavioral deficits in male mice. The mice were divided into normal diet and HFD groups and were analyzed for sleep patterns, behavior tests, and transcription levels of dopamine-related genes in the brain. The HFD group showed decreased wakefulness, increased REM sleep with fragmented patterns, decreased time spent in the center zone of the open field test, shorter immobile time in the tail suspension test, impaired visuospatial memory, and reduced sucrose preference. Additionally, the HFD group had decreased mRNA levels of D1R, COMT, and DAT in the nucleus accumbens, which negatively correlated with REM sleep proportion and REM sleep bout count. The results suggest that HFD-induced behavioral deficits were resemblance to ADHD-like behavioral phenotypes and disturbs REM sleep by dysregulating the dopaminergic system.
Collapse
Affiliation(s)
- Jiseung Kang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Mincheol Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| | - Tae Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| |
Collapse
|
15
|
He Y, Wang Q, Wu H, Dong Y, Peng Z, Guo X, Jiang N. The role of IGF-1 in exercise to improve obesity-related cognitive dysfunction. Front Neurosci 2023; 17:1229165. [PMID: 37638322 PMCID: PMC10447980 DOI: 10.3389/fnins.2023.1229165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/31/2023] [Indexed: 08/29/2023] Open
Abstract
Obesity is an important factor that threatens human health. The occurrence of many chronic diseases is related to obesity, and cognitive function decline often occurs with the onset of obesity. With the further prevalence of obesity, it is bound to lead to a wider range of cognitive dysfunction (ORCD). Therefore, it is crucial to suppress ORCD through intervention. In this regard, exercise has been shown to be effective in preventing obesity and improving cognitive function as a non-drug treatment. There is sufficient evidence that exercise has a regulatory effect on a growth factor closely related to cognitive function-insulin-like growth factor 1 (IGF-1). IGF-1 may be an important mediator in improving ORCD through exercise. This article reviews the effects of obesity and IGF-1 on cognitive function and the regulation of exercise on IGF-1. It analyzes the mechanism by which exercise can improve ORCD by regulating IGF-1. Overall, this review provides evidence from relevant animal studies and human studies, showing that exercise plays a role in improving ORCD. It emphasizes the importance of IGF-1, which helps to understand the health effects of exercise and promotes research on the treatment of ORCD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ning Jiang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise and Health, Tianjin University of Sport, Tianjin, China
| |
Collapse
|
16
|
Chakraborty N, Gautam A, Muhie S, Miller SA, Meyerhoff J, Sowe B, Jett M, Hammamieh R. Potential roles of polyunsaturated fatty acid-enriched diets in modulating social stress-like features. J Nutr Biochem 2023; 116:109309. [PMID: 36871836 DOI: 10.1016/j.jnutbio.2023.109309] [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: 10/13/2022] [Revised: 01/13/2023] [Accepted: 02/24/2023] [Indexed: 03/07/2023]
Abstract
Fish oil or its major constituents, namely omega-3 poly-unsaturated fatty acid (n3-PUFA), are popular supplements to improve neurogenesis, neuroprotection, and overall brain functions. Our objective was to probe the implications of fat enriched diet with variable PUFAs supplements in ameliorating social stress (SS). We fed mice on either of the three diet types, namely the n-3 PUFA-enriched diet (ERD, n3:n6= 7:1), a balanced diet (BLD, n3:n6= 1:1) or a standard lab diet (STD, n3:n6= 1:6). With respect to the gross fat contents, the customized special diets, namely ERD and BLD were extreme diet, not reflecting the typical human dietary composition. Aggressor-exposed SS (Agg-E SS) model triggered behavioral deficiencies that lingered for 6 weeks (6w) post-stress in mice on STD. ERD and BLD elevated bodyweights but potentially helped in building the behavioral resilience to SS. STD adversely affected the gene networks of brain transcriptomics associated with the cell mortality, energy homeostasis and neurodevelopment disorder. Diverging from the ERD's influences on these networks, BLD showed potential long-term benefits in combatting Agg-E SS. The gene networks linked to cell mortality and energy homeostasis, and their subfamilies, such as cerebral disorder and obesity remained at the baseline level of Agg-E SS mice on BLD 6w post-stress. Moreover, neurodevelopment disorder network and its subfamilies like behavioral deficits remained inhibited in the cohort fed on BLD 6w post Agg-E SS.
Collapse
Affiliation(s)
- Nabarun Chakraborty
- Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
| | - Aarti Gautam
- Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Seid Muhie
- Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA; Geneva Foundation, Silver Spring, Maryland, USA
| | - Stacy-Ann Miller
- Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - James Meyerhoff
- Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA; Geneva Foundation, Silver Spring, Maryland, USA
| | - Bintu Sowe
- Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA; Geneva Foundation, Silver Spring, Maryland, USA
| | - Marti Jett
- Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Rasha Hammamieh
- Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| |
Collapse
|
17
|
Vilela WR, Bellozi PMQ, Picolo VL, Cavadas BN, Marques KVS, Pereira LTG, Guirao ARDY, Amato AA, Magalhães KG, Mortari MR, Medei EH, Goulart JT, de Bem AF. Early-life metabolic dysfunction impairs cognition and mitochondrial function in mice. J Nutr Biochem 2023; 117:109352. [PMID: 37061011 DOI: 10.1016/j.jnutbio.2023.109352] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 03/10/2023] [Accepted: 04/10/2023] [Indexed: 04/17/2023]
Abstract
The impact of overnutrition early in life is not restricted to the onset of cardiovascular and metabolic diseases, but also affects critical brain functions related to cognition. This study aimed to evaluate the relationship between peripheral metabolic and bioenergetic changes induced by a two-hit protocol and their impact on cognitive function in juvenile mice. Three-week-old male C57BL/6 mice received a high-fat diet (HFD) or control diet for 7 weeks, associated with 2 low doses of streptozotocin (STZ) or vehicle. Despite the absence of obesity, HFD+STZ impaired glucose metabolism and induced a trend towards cholesterol increase. The two-hit protocol impaired recognition and spatial memories in juvenile mice, without inducing a depressive-like behavior. HFD+STZ mice presented increased immunoreactivity for GFAP and a trend towards a decrease in NeuN in the hippocampus. The treatment caused a bioenergetic impairment in the hippocampus, characterized by a decrease in both O2 consumption related to ATP production and in the maximum respiratory capacity. The thermogenic capacity of brown adipose tissue was impaired by the two-hit protocol, here verified through the absence of a decrease in O2 consumption after uncoupled protein-1 inhibition and an increase in the reserve respiratory capacity. Impaired mitochondrial function was also observed in the liver of HFD+STZ juvenile mice, but not in their heart. These results indicate that exposure to HFD+STZ early in life has a detrimental impact on the bioenergetic and mitochondrial function of tissues with metabolic and thermogenic activities, which is likely related to hippocampal metabolic changes and cognitive impairment.
Collapse
Affiliation(s)
- Wembley Rodrigues Vilela
- Laboratory of Bioenergetics and Metabolism, Department of Physiological Sciences, Biology Institute, University of Brasilia, Federal District, Brazil
| | - Paula Maria Quaglio Bellozi
- Laboratory of Bioenergetics and Metabolism, Department of Physiological Sciences, Biology Institute, University of Brasilia, Federal District, Brazil
| | - Victor Luna Picolo
- Laboratory of Bioenergetics and Metabolism, Department of Physiological Sciences, Biology Institute, University of Brasilia, Federal District, Brazil
| | - Bruna Neves Cavadas
- Laboratory of Bioenergetics and Metabolism, Department of Physiological Sciences, Biology Institute, University of Brasilia, Federal District, Brazil
| | - Keila Valentina Silva Marques
- Laboratory of Bioenergetics and Metabolism, Department of Physiological Sciences, Biology Institute, University of Brasilia, Federal District, Brazil
| | | | - Ainhoa Rodriguez de Yurre Guirao
- Laboratory of Cardioimunology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Angélica Amorim Amato
- Laboratory of Molecular Pharmacology, School of Health Sciences, University of Brasilia, Brazil
| | - Kelly Grace Magalhães
- Laboratory of Immunology and Inflammation, Department of Physiological Sciences, Biology Institute, University of Brasilia, Federal District, Brazil
| | - Márcia Renata Mortari
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Biology Institute, University of Brasilia, Federal District, Brazil
| | - Emiliano Horacio Medei
- Laboratory of Cardioimunology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jair Trapé Goulart
- Laboratory of Bioenergetics and Metabolism, Department of Physiological Sciences, Biology Institute, University of Brasilia, Federal District, Brazil.
| | - Andreza Fabro de Bem
- Laboratory of Bioenergetics and Metabolism, Department of Physiological Sciences, Biology Institute, University of Brasilia, Federal District, Brazil; Center of Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Faculty of Medicine and Health, Linköping University, Linköping, Sweden.
| |
Collapse
|
18
|
Wei W, Lin Z, Xu P, Lv X, Lin L, Li Y, Zhou Y, Lu T, Xue X. Diet Control and Swimming Exercise Ameliorate HFD-Induced Cognitive Impairment Related to the SIRT1-NF- κB/PGC-1 α Pathways in ApoE-/- Mice. Neural Plast 2023; 2023:9206875. [PMID: 36999158 PMCID: PMC10049848 DOI: 10.1155/2023/9206875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/04/2023] [Accepted: 03/07/2023] [Indexed: 04/01/2023] Open
Abstract
High-fat diet- (HFD-) induced neuroinflammation may ultimately lead to an increased risk of cognitive impairment. Here, we evaluate the effects of diet control and swimming or both on the prevention of cognitive impairment by enhancing SIRT1 activity. Twenty-week-old ApoE-/- mice were fed a HFD for 8 weeks and then were treated with diet control and/or swimming for 8 weeks. Cognitive function was assessed using the novel object recognition test (NORT) and Y-maze test. The expression of sirtuin-1 (SIRT1), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), brain-derived neurotrophic factor (BDNF), nuclear factor kappa B p65 (NF-κB p65), interleukin-1β (IL-1β), and tumour necrosis factor-α (TNF-α) in the hippocampus was measured by western blotting. The levels of fractional anisotropy (FA), N-acetylaspartate (NAA)/creatine (Cr) ratio, choline (Cho)/Cr ratio, and myo-inositol (MI)/Cr ratio in the hippocampus were evaluated by diffusion tensor imaging (DTI) and magnetic resonance spectroscopy (MRS) using 7.0-T magnetic resonance imaging (MRI). Our results showed that cognitive dysfunction and hippocampal neuroinflammation appeared to be remarkably observed in apolipoprotein E (ApoE)-/- mice fed with HFD. Diet control plus swimming significantly reversed HFD-induced cognitive decline, reduced the time spent exploring the novel object, and ameliorated spontaneous alternation in the Y-maze test. Compared with the HFD group, ApoE-/- mice fed diet control and/or subjected to swimming had an increase in FA, NAA/Cr, and Cho/Cr; a drop in MI/Cr; elevated expression levels of SIRT1, PGC-1α, and BDNF; and inhibited production of proinflammatory cytokines, including NF-κB p65, IL-1β, and TNF-α. SIRT1, an NAD+-dependent class III histone enzyme, deacetylases and regulates the activity of PGC-1α and NF-κB. These data indicated that diet control and/or swimming ameliorate cognitive deficits through the inhibitory effect of neuroinflammation via SIRT1-mediated pathways, strongly suggesting that swimming and/or diet control could be potentially effective nonpharmacological treatments for cognitive impairment.
Collapse
Affiliation(s)
- Wei Wei
- 1The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Zhicheng Lin
- 1The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - PeiTao Xu
- 1The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xinru Lv
- 2College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Libin Lin
- 2College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yongxu Li
- 2College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yangjie Zhou
- 2College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Taotao Lu
- 2College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xiehua Xue
- 1The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- 3Fujian Provincial Rehabilitation Industrial Institution, Fujian Provincial Key Laboratory of Rehabilitation Technology, Fujian Key Laboratory of Cognitive Rehabilitation, Fuzhou, China
| |
Collapse
|
19
|
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
|
20
|
Swimming Suppresses Cognitive Decline of HFD-Induced Obese Mice through Reversing Hippocampal Inflammation, Insulin Resistance, and BDNF Level. Nutrients 2022; 14:nu14122432. [PMID: 35745162 PMCID: PMC9228449 DOI: 10.3390/nu14122432] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/04/2022] [Accepted: 06/06/2022] [Indexed: 02/01/2023] Open
Abstract
Obesity is an important public health problem nowadays. Long-term obesity can trigger a series of chronic diseases and impair the learning and memory function of the brain. Current studies show that scientific exercise can effectively improve learning and memory capacity, which also can provide benefits for obese people. However, the underlying mechanisms for the improvement of cognitive capacity under the status of obesity still need to be further explored. In the present study, the obesity-induced cognition-declined model was established using 4-week-old mice continuously fed with a high-fat diet (HFD) for 12 weeks, and then the model mice were subjected to an 8-week swimming intervention and corresponding evaluation of relevant indicators, including cognitive capacity, inflammation, insulin signal pathway, brain-derived neurotrophic factor (BNDF), and apoptosis, for exploring potential regulatory mechanisms. Compared with the mice fed with regular diets, the obese mice revealed the impairment of cognitive capacity; in contrast, swimming intervention ameliorated the decline in cognitive capacity of obese mice by reducing inflammatory factors, inhibiting the JNK/IRS-1/PI3K/Akt signal pathway, and activating the PGC-1α/BDNF signal pathway, thereby suppressing the apoptosis of neurons. Therefore, swimming may be an important interventional strategy to compensate for obesity-induced cognitive impairment.
Collapse
|
21
|
Obesity: The Fat Tissue Disease Version of Cancer. Cells 2022; 11:cells11121872. [PMID: 35741001 PMCID: PMC9221301 DOI: 10.3390/cells11121872] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 02/06/2023] Open
Abstract
Obesity is a disease with high potential for fatality. It perfectly fits the disease definition, as cancer does. This is because it damages body structure and functions, both mechanically and biologically, and alters physical, mental, and social health. In addition, it shares many common morbid characteristics with the most feared disease, cancer. For example, it is influenced by a sophisticated interaction between a person’s genetics, the environment, and an increasing number of other backgrounds. Furthermore, it displays abnormal cell growth and proliferation events, only limited to white fat, resulting in adipose tissue taking up an increasing amount of space within the body. This occurs through fat “metastases” and via altered signaling that further aggravates the pathology of obesity by inducing ubiquitous dishomeostasis. These metastases can be made graver by angiogenesis, which might boost diseased tissue growth. More common features with cancer include its progressive escalation through different levels of severity and its possibility of re-onset after recovery. Despite all these similarities with cancer, obesity is substantially less agitating for most people. Thus, the ideas proposed herein could have utility to sensitize the public opinion about the hard reality of obesity. This is increasingly needed, as the obesity pandemic has waged a fierce war against our bodies and society in general, while there is still doubt about whether it is a real disease or not. Hence, raising public consciousness to properly face health issues is crucial to improving our health instead of gaining weight unhealthily. It is obviously illogical to fight cancer extremely seriously on the one hand and to consider dying with obesity as self-inflicted on the other. In fact, obesity merits a top position among the most lethal diseases besides cancer.
Collapse
|
22
|
Thomson S, Chan YL, Yi C, Wang B, Machaalani R, Oliver B, Gorrie CA, Chen H. The impact of high fat consumption on neurological functions following a traumatic brain injury in rats. J Neurotrauma 2022; 39:1547-1560. [PMID: 35658673 DOI: 10.1089/neu.2022.0080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) and obesity are two common conditions in modern society; both can impair neuronal integrity and neurological function. However, it is unclear whether the co-existence of both conditions will worsen outcomes. Thus, in a rat model, we aimed to investigate whether the co-existence of TBI and a high-fat diet (HFD) has an additive effect, leading to more severe neurological impairments, and whether they are related to changes in brain protein markers of oxidative stress, inflammation and synaptic plasticity. Sprague-Dawley rats (female, ~250g) were divided into HFD (43% fat) and chow diet (CD, 17% fat) groups for 6 weeks. Within each dietary group, half underwent a TBI by a weight-drop device, and the other half underwent sham surgery. Short-term memory and sensory function were measured at 24 hours, 1 week, 3 weeks and 6 weeks post-TBI. Brain tissues were harvested at 24 hours and 6 weeks post-TBI and markers of oxidative stress, apoptosis, inflammation, and synaptic plasticity were measured via immunostaining and western blotting. In rats without TBI, HFD increased the presynaptic protein synaptophysin. In rats with TBI, HFD resulted in worsened sensory and memory function, an increase in activated macrophages, and a decrease in the endogenous antioxidant manganese superoxide dismutase. Our findings suggest that the additive effect of HFD and TBI worsens short term memory and sensation deficits, and may be driven by enhanced oxidative stress and inflammation.
Collapse
Affiliation(s)
- Shannon Thomson
- University of Technology Sydney, 1994, School of Life Sciences, Faculty of Science , Sydney, New South Wales, Australia;
| | - Yik Lung Chan
- University of Technology Sydney, 1994, School of Life Sciences, Faculty of Science , Sydney, New South Wales, Australia;
| | - Chenju Yi
- The Seventh Affiliated Hospital Sun Yat-sen University, 543160, 628 Zhenyuan Road, Guangming 518107 Shenzhen China, Shenzhen, China, 518107;
| | - Baoming Wang
- University of Technology Sydney, 1994, School of Life Sciences, Faculty of Science , Sydney, New South Wales, Australia;
| | - Rita Machaalani
- University of Technology Sydney, 1994, Faculty of Medicine and Health, Sydney, New South Wales, Australia;
| | - Brian Oliver
- University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, New South Wales, Australia.,The University of Sydney, RCMB, Woolcock Institute of Medical Research, Sydney, New South Wales, Australia;
| | - Catherine A Gorrie
- University of Technology Sydney, School of Life Sciences, Faculty of Science, PO Box 123, Broadway, Sydney, New South Wales, Australia, 2007;
| | - Hui Chen
- University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia;
| |
Collapse
|
23
|
Park SK, Lee HL, Kang JY, Kim JM, Heo HJ. Peanut (Arachis hypogaea) sprout prevents high-fat diet-induced cognitive impairment by improving mitochondrial function. Sci Rep 2022; 12:6213. [PMID: 35418581 PMCID: PMC9008020 DOI: 10.1038/s41598-022-10520-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 04/06/2022] [Indexed: 11/09/2022] Open
Abstract
This study was performed to evaluate the improvement effect of the ethyl acetate fraction from peanut (Arachis hypogaea) sprout (EFPS) on high-fat diet (HFD)-induced cognitive deficits in C57BL/6 mice. Mice were randomly divided four groups (n = 13) as control (normal chow), HFD, EFPS 20 (20 mg/kg of body weight; intragastric administration) and EFPS 50 (50 mg/kg of body weight; intragastric administration) groups. HFD was provide for 15 weeks excepting control group. EFPS ameliorated cognitive dysfunction in Y-maze, passive avoidance test and Morris water maze test. EFPS significantly improved glucose tolerance and serum lipid profile, and reduced body weight. EFPS ameliorated oxidative stress by regulating MDA levels and SOD activity in liver and brain tissues. In addition, EFPS restored brain mitochondrial dysfunction related to energy metabolism. Moreover, the bioactive compounds of EFPS were identified as di-caffeic acid, caffeic acid, dihydrokaempferol-hexoside, di-p-coumaroyl tartaric acid isomer and group B soyasaponins using ultra-performance liquid chromatography-quadrupole-time-of-flight (UPLC-Q-TOF) mass spectrometry. These results show that EFPS can improve cognitive functions in HFD-induced diabetic mice.
Collapse
Affiliation(s)
- Seon Kyeong Park
- Division of Applied Life Science, Institute of Agriculture and Life Science (BK21), Gyeongsang National University, Jinju, 52828, Republic of Korea
- Korea Food Research Institute, Wanju-gun, 55365, Republic of Korea
| | - Hyo Lim Lee
- Division of Applied Life Science, Institute of Agriculture and Life Science (BK21), Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Jin Yong Kang
- Division of Applied Life Science, Institute of Agriculture and Life Science (BK21), Gyeongsang National University, Jinju, 52828, Republic of Korea
- Advanced Process Technology and Fermentation Research Group, World Institute of Kimchi, Gwangju, 61755, Republic of Korea
| | - Jong Min Kim
- Division of Applied Life Science, Institute of Agriculture and Life Science (BK21), Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Ho Jin Heo
- Division of Applied Life Science, Institute of Agriculture and Life Science (BK21), Gyeongsang National University, Jinju, 52828, Republic of Korea.
| |
Collapse
|
24
|
Gupta R, Sahu M, Tripathi R, Ambasta RK, Kumar P. Protein S-sulfhydration: Unraveling the prospective of hydrogen sulfide in the brain, vasculature and neurological manifestations. Ageing Res Rev 2022; 76:101579. [PMID: 35124235 DOI: 10.1016/j.arr.2022.101579] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/30/2022] [Accepted: 02/01/2022] [Indexed: 02/08/2023]
Abstract
Hydrogen sulfide (H2S) and hydrogen polysulfides (H2Sn) are essential regulatory signaling molecules generated by the entire body, including the central nervous system. Researchers have focused on the classical H2S signaling from the past several decades, whereas the last decade has shown the emergence of H2S-induced protein S-sulfhydration signaling as a potential therapeutic approach. Cysteine S-persulfidation is a critical paradigm of post-translational modification in the process of H2S signaling. Additionally, studies have shown the cross-relationship between S-sulfhydration and other cysteine-induced post-translational modifications, namely nitrosylation and carbonylation. In the central nervous system, S-sulfhydration is involved in the cytoprotection through various signaling pathways, viz. inflammatory response, oxidative stress, endoplasmic reticulum stress, atherosclerosis, thrombosis, and angiogenesis. Further, studies have demonstrated H2S-induced S-sulfhydration in regulating different biological processes, such as mitochondrial integrity, calcium homeostasis, blood-brain permeability, cerebral blood flow, and long-term potentiation. Thus, protein S-sulfhydration becomes a crucial regulatory molecule in cerebrovascular and neurodegenerative diseases. Herein, we first described the generation of intracellular H2S followed by the application of H2S in the regulation of cerebral blood flow and blood-brain permeability. Further, we described the involvement of S-sulfhydration in different biological and cellular functions, such as inflammatory response, mitochondrial integrity, calcium imbalance, and oxidative stress. Moreover, we highlighted the importance of S-sulfhydration in cerebrovascular and neurodegenerative diseases.
Collapse
|
25
|
Leal PEDPT, da Silva AA, Rocha-Gomes A, Riul TR, Cunha RA, Reichetzeder C, Villela DC. High-Salt Diet in the Pre- and Postweaning Periods Leads to Amygdala Oxidative Stress and Changes in Locomotion and Anxiety-Like Behaviors of Male Wistar Rats. Front Behav Neurosci 2022; 15:779080. [PMID: 35058757 PMCID: PMC8763963 DOI: 10.3389/fnbeh.2021.779080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/15/2021] [Indexed: 12/24/2022] Open
Abstract
High-salt (HS) diets have recently been linked to oxidative stress in the brain, a fact that may be a precursor to behavioral changes, such as those involving anxiety-like behavior. However, to the best of our knowledge, no study has evaluated the amygdala redox status after consuming a HS diet in the pre- or postweaning periods. This study aimed to evaluate the amygdala redox status and anxiety-like behaviors in adulthood, after inclusion of HS diet in two periods: preconception, gestation, and lactation (preweaning); and only after weaning (postweaning). Initially, 18 females and 9 male Wistar rats received a standard (n = 9 females and 4 males) or a HS diet (n = 9 females and 5 males) for 120 days. After mating, females continued to receive the aforementioned diets during gestation and lactation. Weaning occurred at 21-day-old Wistar rats and the male offspring were subdivided: control-control (C-C)—offspring of standard diet fed dams who received a standard diet after weaning (n = 9–11), control-HS (C-HS)—offspring of standard diet fed dams who received a HS diet after weaning (n = 9–11), HS-C—offspring of HS diet fed dams who received a standard diet after weaning (n = 9–11), and HS-HS—offspring of HS diet fed dams who received a HS diet after weaning (n = 9–11). At adulthood, the male offspring performed the elevated plus maze and open field tests. At 152-day-old Wistar rats, the offspring were euthanized and the amygdala was removed for redox state analysis. The HS-HS group showed higher locomotion and rearing frequency in the open field test. These results indicate that this group developed hyperactivity. The C-HS group had a higher ratio of entries and time spent in the open arms of the elevated plus maze test in addition to a higher head-dipping frequency. These results suggest less anxiety-like behaviors. In the analysis of the redox state, less activity of antioxidant enzymes and higher levels of the thiobarbituric acid reactive substances (TBARS) in the amygdala were shown in the amygdala of animals that received a high-salt diet regardless of the period (pre- or postweaning). In conclusion, the high-salt diet promoted hyperactivity when administered in the pre- and postweaning periods. In animals that received only in the postweaning period, the addition of salt induced a reduction in anxiety-like behaviors. Also, regardless of the period, salt provided amygdala oxidative stress, which may be linked to the observed behaviors.
Collapse
Affiliation(s)
- Pedro Ernesto de Pinho Tavares Leal
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
- Laboratório de Nutrição Experimental – LabNutrex, Departamento de Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Alexandre Alves da Silva
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
- Laboratório de Nutrição Experimental – LabNutrex, Departamento de Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Arthur Rocha-Gomes
- Laboratório de Nutrição Experimental – LabNutrex, Departamento de Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Tania Regina Riul
- Laboratório de Nutrição Experimental – LabNutrex, Departamento de Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
- Programa de Pós-Graduação em Ciências da Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Rennan Augusto Cunha
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Christoph Reichetzeder
- Department of Nutritional Toxicology, Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
- Christoph Reichetzeder,
| | - Daniel Campos Villela
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
- *Correspondence: Daniel Campos Villela,
| |
Collapse
|
26
|
de Paula GC, Brunetta HS, Engel DF, Gaspar JM, Velloso LA, Engblom D, de Oliveira J, de Bem AF. Hippocampal Function Is Impaired by a Short-Term High-Fat Diet in Mice: Increased Blood-Brain Barrier Permeability and Neuroinflammation as Triggering Events. Front Neurosci 2021; 15:734158. [PMID: 34803583 PMCID: PMC8600238 DOI: 10.3389/fnins.2021.734158] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Worldwide, and especially in Western civilizations, most of the staple diets contain high amounts of fat and refined carbohydrates, leading to an increasing number of obese individuals. In addition to inducing metabolic disorders, energy dense food intake has been suggested to impair brain functions such as cognition and mood control. Here we demonstrate an impaired memory function already 3 days after the start of a high-fat diet (HFD) exposure, and depressive-like behavior, in the tail suspension test, after 5 days. These changes were followed by reduced synaptic density, changes in mitochondrial function and astrocyte activation in the hippocampus. Preceding or coinciding with the behavioral changes, we found an induction of the proinflammatory cytokines TNF-α and IL-6 and an increased permeability of the blood–brain barrier (BBB), in the hippocampus. Finally, in mice treated with a TNF-α inhibitor, the behavioral and BBB alterations caused by HFD-feeding were mitigated suggesting that inflammatory signaling was critical for the changes. In summary, our findings suggest that HFD rapidly triggers hippocampal dysfunction associated with BBB disruption and neuroinflammation, promoting a progressive breakdown of synaptic and metabolic function. In addition to elucidating the link between diet and cognitive function, our results might be relevant for the comprehension of the neurodegenerative process.
Collapse
Affiliation(s)
- Gabriela Cristina de Paula
- Postgraduate Program in Biochemistry, Department of Biochemistry, Federal University of Santa Catarina, Florianopolis, Brazil.,Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Henver S Brunetta
- Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Daiane F Engel
- School of Pharmacy, Federal University of Ouro Preto, Ouro Preto, Brazil.,Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Joana M Gaspar
- Postgraduate Program in Biochemistry, Department of Biochemistry, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Licio A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - David Engblom
- Department of Clinical and Experimental Medicine, Faculty of Medicine and Health, Linköping University, Linköping, Sweden
| | - Jade de Oliveira
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Andreza Fabro de Bem
- Department of Clinical and Experimental Medicine, Faculty of Medicine and Health, Linköping University, Linköping, Sweden.,Department of Physiological Science, University of Brasília, Brasília, Brazil
| |
Collapse
|
27
|
Sarangi M, Dus M. Crème de la Créature: Dietary Influences on Behavior in Animal Models. Front Behav Neurosci 2021; 15:746299. [PMID: 34658807 PMCID: PMC8511460 DOI: 10.3389/fnbeh.2021.746299] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/06/2021] [Indexed: 11/13/2022] Open
Abstract
In humans, alterations in cognitive, motivated, and affective behaviors have been described with consumption of processed diets high in refined sugars and saturated fats and with high body mass index, but the causes, mechanisms, and consequences of these changes remain poorly understood. Animal models have provided an opportunity to answer these questions and illuminate the ways in which diet composition, especially high-levels of added sugar and saturated fats, contribute to brain physiology, plasticity, and behavior. Here we review findings from invertebrate (flies) and vertebrate models (rodents, zebrafish) that implicate these diets with changes in multiple behaviors, including eating, learning and memory, and motivation, and discuss limitations, open questions, and future opportunities.
Collapse
Affiliation(s)
| | - Monica Dus
- Department of Molecular, Cellular, and Developmental Biology, The University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
28
|
Ge Q, Hu X, Ma N, Sun M, Zhang L, Cai Z, Tan R, Lu H. Maternal high-salt diet during pregnancy impairs synaptic plasticity and memory in offspring. FASEB J 2021; 35:e21244. [PMID: 33715195 DOI: 10.1096/fj.202001890r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 11/11/2022]
Abstract
Excess salt intake harms the brain health and cognitive functions, but whether a maternal high-salt diet (HSD) affects the brain development and neural plasticity of offspring remains unclear. Here, using a range of behavioral tests, we reported that the offspring of maternal HSD subjects exhibited short- and long-term memory deficits, especially in spatial memory in adulthood. Moreover, impairments in synaptic transmission and plasticity in the hippocampus were observed in adult offspring by using in vivo electrophysiology. Consistently, the number of astrocytes but not neurons in the hippocampus of the offspring from the HSD group were significantly decreased, and ERK and AKT signaling pathways involved in neurodevelopment were highly activated only during juvenile. In addition, the expression of synaptic proteins decreased both in juvenile and adulthood, and this effect might be involved in synaptic dysfunction. Collectively, these data demonstrated that the maternal HSD might cause adult offspring synaptic dysfunction and memory loss. It is possibly due to the reduction of astrocytes in juvenile.
Collapse
Affiliation(s)
- Qian Ge
- Department of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Xiaoxuan Hu
- Department of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China.,Department of Human Anatomy and Histo-embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Ning Ma
- Department of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China.,Department of Human Anatomy and Histo-embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Meiqi Sun
- Department of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Liyun Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China.,Department of Human Anatomy and Histo-embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Zhenlu Cai
- Department of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Ruolan Tan
- Department of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China.,Department of Human Anatomy and Histo-embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Haixia Lu
- Department of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| |
Collapse
|
29
|
Pontifex MG, Martinsen A, Saleh RNM, Harden G, Tejera N, Müller M, Fox C, Vauzour D, Minihane AM. APOE4 genotype exacerbates the impact of menopause on cognition and synaptic plasticity in APOE-TR mice. FASEB J 2021; 35:e21583. [PMID: 33891334 DOI: 10.1096/fj.202002621rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 01/15/2023]
Abstract
The impact of sex and menopausal status in Alzheimer's disease remains understudied despite increasing evidence of greater female risk, particularly in APOE4 carriers. Utilizing female APOE-TR mice maintained on a high-fat diet background we induced ovarian failure through repeated VCD injections, to mimic human menopause. At 12 months of age, recognition memory and spatial memory were assessed using object recognition, Y-maze spontaneous alternation, and Barnes maze. A VCD*genotype interaction reduced the recognition memory (P < .05), with APOE4 VCD-treated animals unable to distinguish between novel and familiar objects. APOE4 mice displayed an additional 37% and 12% reduction in Barnes (P < .01) and Y-maze (P < .01) performance, indicative of genotype-specific spatial memory impairment. Molecular analysis indicated both VCD and genotype-related deficits in synaptic plasticity with BDNF, Akt, mTOR, and ERK signaling compromised. Subsequent reductions in the transcription factors Creb1 and Atf4 were also evident. Furthermore, the VCD*genotype interaction specifically diminished Ephb2 expression, while Fos, and Cnr1 expression reduced as a consequence of APOE4 genotype. Brain DHA levels were 13% lower in VCD-treated animals independent of genotype. Consistent with this, we detected alterations in the expression of the DHA transporters Acsl6 and Fatp4. Our results indicate that the combination of ovarian failure and APOE4 leads to an exacerbation of cognitive and neurological deficits.
Collapse
Affiliation(s)
| | | | | | - Glenn Harden
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Noemi Tejera
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Michael Müller
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Chris Fox
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - David Vauzour
- Norwich Medical School, University of East Anglia, Norwich, UK
| | | |
Collapse
|
30
|
Sharma S. High fat diet and its effects on cognitive health: alterations of neuronal and vascular components of brain. Physiol Behav 2021; 240:113528. [PMID: 34260890 DOI: 10.1016/j.physbeh.2021.113528] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/03/2021] [Accepted: 07/06/2021] [Indexed: 01/01/2023]
Abstract
It has been well recognized that intake of diets rich in saturated fats could result in development of metabolic disorders such as type 2 diabetes mellitus, obesity and cardiovascular diseases. Recent studies have suggested that intake of high fat diet (HFD) is also associated with cognitive dysfunction. Various preclinical studies have demonstrated the impact of short and long term HFD feeding on the biochemical and behavioural alterations. This review summarizes studies and the protocols used to assess the impacts of HFD feeding on cognitive performance in rodents. Further, it discuss the key mechanisms that are altered by HFD feeding, such as, insulin resistance, oxidative stress, neuro-inflammation, transcriptional dysregulation and loss of synaptic plasticity. Along with these, HFD feeding also alters the vascular components of brain such as loss of BBB integrity and reduced cerebral blood flow. It is highly possible that these factors are responsible for the development of cognitive deficits as a result of HFD feeding.
Collapse
Affiliation(s)
- Sorabh Sharma
- Division of Medical Sciences, University of Victoria, PO Box 1700 STN CSC, Victoria, BC, V8W2Y2, Canada.
| |
Collapse
|
31
|
Alkan I, Altunkaynak BZ, Gültekin Gİ, Bayçu C. Hippocampal neural cell loss in high-fat diet-induced obese rats-exploring the protein networks, ultrastructure, biochemical and bioinformatical markers. J Chem Neuroanat 2021; 114:101947. [PMID: 33766576 DOI: 10.1016/j.jchemneu.2021.101947] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/27/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Obesity, which has become one of the main health problems, results from irregular and unhealthy nutrition. In particular, an increase in the intake of high-fat foods leads to obesity and associated disorders. It is noteworthy to specify that obese individuals have memory problems. This study aims to examine the effects of high-fat diet on hippocampus, with stereological, histopathological methods and STRING bioinformatic tool. METHODS Female Adult Sprague Dawley rats (n = 20) were equally divided into control (CONT) and high-fat diet (HFD) groups. The control group was given standard rat pellet feed, while the high-fat diet group was fed with a 40 % fat content for 2 months. Following the feeding program, rats were sacrificed. The collected blood samples were analyzed biochemically to determine the level of oxidative stress while performing a stereological and histopathological examination of the brain tissues. Functional protein-protein networks for BDNF, C-Fos, CAT, LPO, SOD and MPO by gene ontology (GO) enrichment analysis were evaluated. FINDINGS The number of neurons decreased in the HFD group compared to the CONT group. Damage to the histological structure of the hippocampus region; such as degenerate neurons, damaged mitochondria and extended cisterns of the endoplasmic reticulum was observed. Although C-Fos level and oxidative stress parameters increased in HFD group, BDNF level decreased. While BDNF and C-Fos were observed in pathways related to neuron death, oxidative stress and memory, BDNF was pronounced in the mitochondria, and C-Fos in the endoplasmic reticulum. DISCUSSION This study shows that changes in both BDNF and C-Fos levels in obesity due to high-fat diet increase oxidative stress and cause neuron damage in the hippocampus.
Collapse
Affiliation(s)
- Işınsu Alkan
- Dept of Basic Medical Sciences, Dentistry Faculty, Nevşehir Hacı Bektaş Veli University, Nevşehir Turkey
| | - Berrin Zuhal Altunkaynak
- Depts of Histology and Embryology and Physiology Departments, Medical Faculty, Istanbul Okan University, İstanbul, Turkey.
| | - Güldal İnal Gültekin
- Physiology Department, Medical Faculty, Istanbul Okan University, İstanbul, Turkey
| | - Cengiz Bayçu
- Histology Department, Medical Faculty, Istanbul Okan University, İstanbul, Turkey
| |
Collapse
|
32
|
Izquierdo V, Palomera-Ávalos V, Pallàs M, Griñán-Ferré C. Resveratrol Supplementation Attenuates Cognitive and Molecular Alterations under Maternal High-Fat Diet Intake: Epigenetic Inheritance over Generations. Int J Mol Sci 2021; 22:1453. [PMID: 33535619 PMCID: PMC7867164 DOI: 10.3390/ijms22031453] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/20/2021] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
Environmental factors such as maternal high-fat diet (HFD) intake can increase the risk of age-related cognitive decline in adult offspring. Epigenetic mechanisms are a possible link between diet effect and neurodegeneration across generations. Here, we found a significant decrease in triglyceride levels in a high-fat diet with resveratrol (RSV) HFD + RSV group and the offspring. Firstly, we obtained better cognitive performance in HFD+RSV groups and their offspring. Molecularly, a significant increase in DNA methylation (5-mC) levels, as well as increased gene expression of DNA methyltransferase 1 (Dnmt1) and Dnmt3a in HFD + RSV F1 group, were found. Furthermore, a significant increase of N6-Methyladenosine methylation (m6A) levels in HFD+RSV F1, as well as changes in gene expression of its enzymes Methyltransferase like 3 (Mettl3) and FTO alpha-ketoglutarate dependent dioxygenase (Fto) were found. Moreover, we found a decrease in gene expression levels of pro-inflammatory markers such as Interleukin 1β (Il1-β), Interleukin 6 (Il-6), Tumor necrosis factor-α (Tnf-α), C-X-C motif chemokine ligand 10 (Cxcl-10), the pro-inflammatory factors monocyte chemoattractant protein 1 (Mcp-1) and Tumor growth factor-β1 (Tgf-β1) in HFD+RSV and HFD+RSV F1 groups. Moreover, there was increased gene expression of neurotrophins such as Neural growth factor (Ngf), Neurotrophin-3 (Nt3), and its receptors Tropomyosin receptor kinase TrkA and TrkB. Likewise, an increase in protein levels of brain-derived neurotrophic factor (BDNF) and phospho-protein kinase B (p-Akt) in HFD+RSV F1 was found. These results suggest that maternal RSV supplementation under HFD intake prevents cognitive decline in senescence-accelerated mice prone 8 (SAMP8) adult offspring, promoting a reduction in triglycerides and leptin plasma levels, changes in the pro-inflammatory profile, and restoring the epigenetic landscape as well as synaptic plasticity.
Collapse
Affiliation(s)
- Vanesa Izquierdo
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències—Universitat de Barcelona, Avda. Joan XXIII, 27, 08028 Barcelona, Spain; (V.I.); (M.P.)
| | - Verónica Palomera-Ávalos
- Department of Cellular and Molecular Biology, University Center of Biological and Agricultural Sciences, University of Guadalajara, km 15.5 Guadalajara-Nogales highway, 45110 Zapopan, Jalisco, Mexico;
| | - Mercè Pallàs
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències—Universitat de Barcelona, Avda. Joan XXIII, 27, 08028 Barcelona, Spain; (V.I.); (M.P.)
| | - Christian Griñán-Ferré
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències—Universitat de Barcelona, Avda. Joan XXIII, 27, 08028 Barcelona, Spain; (V.I.); (M.P.)
| |
Collapse
|
33
|
Kawamura N, Katsuura G, Yamada-Goto N, Novianti E, Inui A, Asakawa A. Impaired brain fractalkine-CX3CR1 signaling is implicated in cognitive dysfunction in diet-induced obese mice. BMJ Open Diabetes Res Care 2021; 9:9/1/e001492. [PMID: 33568358 PMCID: PMC7878130 DOI: 10.1136/bmjdrc-2020-001492] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 12/09/2020] [Accepted: 01/09/2021] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION A diet high in saturated fat is well known to affect neuronal function and contribute to cognitive decline in experimental animals and humans. Fractalkine released from neurons acts on its receptor, CX3C chemokine receptor 1 (CX3CR1), in the microglia to regulate several brain functions. The present study addressed whether fractalkine-CX3CR1 signaling in the brain, especially the hippocampus, contributes to the cognitive deficits observed in diet-induced obese (DIO) mice. RESEARCH DESIGN AND METHODS Mice were given 60% high-fat diet for 16 weeks. The expression of fractalkine and CX3CR1 in the hippocampus, amygdala and prefrontal cortex of DIO mice was analyzed. Cognitive ability in the Y-maze test and hippocampal glutamate receptors and synaptic markers were observed in DIO and CX3CR1 antagonist-treated mice. Regulation of fractalkine and CX3CR1 expression in the hippocampus was examined following administration of a selective insulin-like growth factor-1 (IGF-1) receptor inhibitor and a tyrosine receptor kinase B (TrkB) antagonist in normal mice. RESULTS DIO mice exhibited significant cognitive deficits in the Y-maze test and decrease in fractalkine and CX3CR1 in the hippocampus and amygdala compared with mice fed a control diet (CD mice). Administration of the CX3CR1 antagonist 18a in normal mice induced significant cognitive deficits in the Y-maze test. DIO mice and CX3CR1 antagonist-treated mice exhibited significant decreases in protein levels of NMDA (N-methyl-D-aspartate) receptor subunit (NR2A), AMPA (α-amino-5-methyl-3-hydroxy-4-isoxazole propionate) receptor subunit (GluR1) and postsynaptic density protein 95 in the hippocampus compared with their respective controls. Furthermore, plasma IGF-1 and hippocampal brain-derived neurotrophic factor were significantly decreased in DIO mice compared with CD mice. Administration of a selective IGF-1 receptor inhibitor and a TrkB antagonist in normal mice significantly decreased fractalkine and CX3CR1 in the hippocampus. CONCLUSIONS These findings indicate that the cognitive decline observed in DIO mice is due, in part, to reduced fractalkine-CX3CR1 signaling in the corticolimbic system.
Collapse
Affiliation(s)
- Namiko Kawamura
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Goro Katsuura
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Nobuko Yamada-Goto
- Health Center, Keio University, Shinjuku-ku, Tokyo, Japan
- Division of Endocrinology, Metabolism and Nephrology, Department of Internal Medicine, Keio University, School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Ela Novianti
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Akio Inui
- Pharmacological Department of Herbal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Akihiro Asakawa
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
34
|
From Obesity to Hippocampal Neurodegeneration: Pathogenesis and Non-Pharmacological Interventions. Int J Mol Sci 2020; 22:ijms22010201. [PMID: 33379163 PMCID: PMC7796248 DOI: 10.3390/ijms22010201] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 12/16/2022] Open
Abstract
High-caloric diet and physical inactivity predispose individuals to obesity and diabetes, which are risk factors of hippocampal neurodegeneration and cognitive deficits. Along with the adipose-hippocampus crosstalk, chronically inflamed adipose tissue secretes inflammatory cytokine could trigger neuroinflammatory responses in the hippocampus, and in turn, impairs hippocampal neuroplasticity under obese and diabetic conditions. Hence, caloric restriction and physical exercise are critical non-pharmacological interventions to halt the pathogenesis from obesity to hippocampal neurodegeneration. In response to physical exercise, peripheral organs, including the adipose tissue, skeletal muscles, and liver, can secret numerous exerkines, which bring beneficial effects to metabolic and brain health. In this review, we summarized how chronic inflammation in adipose tissue could trigger neuroinflammation and hippocampal impairment, which potentially contribute to cognitive deficits in obese and diabetic conditions. We also discussed the potential mechanisms underlying the neurotrophic and neuroprotective effects of caloric restriction and physical exercise by counteracting neuroinflammation, plasticity deficits, and cognitive impairments. This review provides timely insights into how chronic metabolic disorders, like obesity, could impair brain health and cognitive functions in later life.
Collapse
|
35
|
Glushchak K, Ficarro A, Schoenfeld TJ. High-fat diet and acute stress have different effects on object preference tests in rats during adolescence and adulthood. Behav Brain Res 2020; 399:112993. [PMID: 33152318 DOI: 10.1016/j.bbr.2020.112993] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/30/2020] [Accepted: 10/28/2020] [Indexed: 12/16/2022]
Abstract
Meals of high-fat diet (HFD) during adolescence produce stronger impairments to memory during adolescence than adulthood, however recovery of memory from adolescent HFD is underexplored. In addition, many tests of rodent memory are confounded by aversive or food-based stimuli, making it difficult to determine baseline memory processing affected by HFD. Thus, we utilized three cohorts of rats (adolescent HFD, adult HFD, and adolescent HFD with recovery) to explore the effects of HFD at different ages on two traditional tests of memory based strictly on object exploration, novel object recognition and novel object location tests. To isolate stress as a variable, rats were tested either at baseline or with cold water swim occurring directly after object acquisition. Results show that preference for novel objects is impaired by stress across all groups, but HFD alone only impairs preference for novel objects during adolescence, although this recovers after switching to a control diet. Additionally, preference for an object in a new location is impaired by HFD in all age groups and fails to recover following diet change. Together the data suggest that stress and HFD differentially affect object preference, based on test type, except during the adolescent period. Because these tests are traditionally interpreted as memory processes dependent on two distinct brain regions, the hippocampus and perirhinal cortex, these results support that stress and HFD affect the hippocampus and perirhinal cortex differently. The data affirm that while perirhinal cortex-dependent behavior recovers, the adolescent period is susceptible to long-lasting dysfunctions of hippocampal behavior by HFD.
Collapse
Affiliation(s)
- Karina Glushchak
- Department of Psychological Science and Neuroscience, Belmont University, Nashville, TN, 37212, USA
| | - Alexandria Ficarro
- Department of Psychological Science and Neuroscience, Belmont University, Nashville, TN, 37212, USA
| | - Timothy J Schoenfeld
- Department of Psychological Science and Neuroscience, Belmont University, Nashville, TN, 37212, USA.
| |
Collapse
|
36
|
Thapak P, Bishnoi M, Sharma SS. Pharmacological Inhibition of Transient Receptor Potential Melastatin 2 (TRPM2) Channels Attenuates Diabetes-induced Cognitive Deficits in Rats: A Mechanistic Study. Curr Neurovasc Res 2020; 17:249-258. [DOI: 10.2174/1567202617666200415142211] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/02/2020] [Accepted: 03/07/2020] [Indexed: 02/08/2023]
Abstract
Background:
Diabetes is a chronic metabolic disorder affecting the central nervous system.
A growing body of evidence has depicted that high glucose level leads to the activation of the
transient receptor potential melastatin 2 (TRPM2) channels. However, there are no studies targeting
TRPM2 channels in diabetes-induced cognitive decline using a pharmacological approach.
Objective:
The present study intended to investigate the effects of 2-aminoethoxydiphenyl borate
(2-APB), a TRPM2 inhibitor, in diabetes-induced cognitive impairment.
Methods:
Streptozotocin (STZ, 50 mg/kg, i.p.) was used to induce diabetes in rats. Animals were
randomly divided into the treatment group, model group and age-matched control and pre se
group. 2-APB treatment was given for three weeks to the animals. After 10 days of behavioural
treatment, parameters were performed. Animals were sacrificed at 10th week of diabetic induction
and the hippocampus and cortex were isolated. After that, protein and mRNA expression study
was performed in the hippocampus. Acetylcholinesterase (AchE) activity was done in the cortex.
Results: :
Our study showed the 10th week diabetic animals developed cognitive impairment, which
was evident from the behavioural parameters. Diabetic animals depicted an increase in the TRPM2
mRNA and protein expression in the hippocampus as well as increased AchE activity in the cortex.
However, memory associated proteins were down-regulated, namely Ca2+/calmodulin-dependent
protein kinase II (CaMKII-Thr286), glycogen synthase kinase 3 beta (GSK-3β-Ser9), cAMP
response element-binding protein (CREB-Ser133), and postsynaptic density protein 95 (PSD-95).
Gene expression of parvalbumin, calsequestrin and brain-derived neurotrophic factor (BDNF)
were down-regulated while mRNA level of calcineurin A/ protein phosphatase 3 catalytic subunit
alpha (PPP3CA) was upregulated in the hippocampus of diabetic animals. A three-week treatment
with 2-APB significantly ameliorated the alteration in behavioural cognitive parameters in diabetic
rats. Moreover, 2-APB also down-regulated the expression of TRPM2 mRNA and protein in the
hippocampus as well as AchE activity in the cortex of diabetic animals as compared to diabetic
animals. Moreover, the 2-APB treatment also upregulated the CaMKII (Thr-286), GSK-3β (Ser9),
CREB (Ser133), and PSD-95 expression and mRNA levels of parvalbumin, calsequestrin, and
BDNF while mRNA level of calcineurin A was down-regulated in the hippocampus of diabetic
animals.
Conclusion: :
This study confirms the ameliorative effect of TRPM2 channel inhibitor in the diabetes-
induced cognitive deficits. Inhibition of TRPM2 channels reduced the calcium associated
downstream signaling and showed a neuroprotective effect of TRPM2 channels in diabetesinduced
cognitive impairment.
Collapse
Affiliation(s)
- Pavan Thapak
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, India
| | - Mahendra Bishnoi
- National Agri-Food Biotechnology Institute, Sector 81, S.A.S. Nagar, Punjab, India
| | - Shyam S. Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, India
| |
Collapse
|
37
|
Interplay between Peripheral and Central Inflammation in Obesity-Promoted Disorders: The Impact on Synaptic Mitochondrial Functions. Int J Mol Sci 2020; 21:ijms21175964. [PMID: 32825115 PMCID: PMC7504224 DOI: 10.3390/ijms21175964] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/12/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022] Open
Abstract
The metabolic dysfunctions induced by high fat diet (HFD) consumption are not limited to organs involved in energy metabolism but cause also a chronic low-grade systemic inflammation that affects the whole body including the central nervous system. The brain has been considered for a long time to be protected from systemic inflammation by the blood–brain barrier, but more recent data indicated an association between obesity and neurodegeneration. Moreover, obesity-related consequences, such as insulin and leptin resistance, mitochondrial dysfunction and reactive oxygen species (ROS) production, may anticipate and accelerate the physiological aging processes characterized by systemic inflammation and higher susceptibility to neurological disorders. Here, we discussed the link between obesity-related metabolic dysfunctions and neuroinflammation, with particular attention to molecules regulating the interplay between energetic impairment and altered synaptic plasticity, for instance AMP-activated protein kinase (AMPK) and Brain-derived neurotrophic factor (BDNF). The effects of HFD-induced neuroinflammation on neuronal plasticity may be mediated by altered brain mitochondrial functions. Since mitochondria play a key role in synaptic areas, providing energy to support synaptic plasticity and controlling ROS production, the negative effects of HFD may be more pronounced in synapses. In conclusion, it will be emphasized how HFD-induced metabolic alterations, systemic inflammation, oxidative stress, neuroinflammation and impaired brain plasticity are tightly interconnected processes, implicated in the pathogenesis of neurological diseases.
Collapse
|
38
|
Prom-in S, Kaewsrichan J, Wangpradit N, Kien Hui C, Yahaya MF, Kamisah Y, Kumar J. Abelmoschus Esculentus (L.) Moench's Peel Powder Improves High-Fat-Diet-Induced Cognitive Impairment in C57BL/6J Mice. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E5513. [PMID: 32751614 PMCID: PMC7432850 DOI: 10.3390/ijerph17155513] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/24/2020] [Accepted: 07/29/2020] [Indexed: 02/08/2023]
Abstract
Okra peel exhibits numerous therapeutic effects. This study explores the potential ameliorative effects of okra peel powder on high-fat-diet (HFD)-induced hypercholesterolemia and cognitive deficits. Thirty-six C57BL/6J male mice were randomly divided into six groups (n = 6 per group): (i) control, mice fed with a normal diet; (ii) HFD, mice fed with HFD; (iii) HFD-SIM, mice fed with HFD and given simvastatin (20 mg/kg/day); (iv) HFD-OP1; (v) HFD-OP2; (vi) HFD-OP3, mice fed with HFD and okra peel (200, 400, or 800 mg/kg/day, respectively). Following 10 weeks of treatments, the mice were subjected to the Morris water maze (MWM). Parameters such as weekly average body weight, food intake, and blood lipid profiles were also recorded. The HFD group showed a profound increase in total cholesterol and low-density lipoprotein concentration compared to the control group. All okra-treated and HFD-SIM groups performed better than the HFD group during acquisition trials, whereas only the HFD-OP1 produced a significantly higher number of entries into the platform zone during the probe trial. In sum, all three okra doses improved the learning ability of the mice. However, only the lowest dose of okra significantly improved the spatial reference memory retention.
Collapse
Affiliation(s)
- Supattra Prom-in
- Department of Pharmaceutical Chemistry and Drug Delivery System Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai, Songkhla 90110, Thailand; (S.P.-i.); (J.K.); (N.W.)
| | - Jasadee Kaewsrichan
- Department of Pharmaceutical Chemistry and Drug Delivery System Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai, Songkhla 90110, Thailand; (S.P.-i.); (J.K.); (N.W.)
| | - Nuntika Wangpradit
- Department of Pharmaceutical Chemistry and Drug Delivery System Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai, Songkhla 90110, Thailand; (S.P.-i.); (J.K.); (N.W.)
| | - Chua Kien Hui
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Mohamad Fairuz Yahaya
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Yusof Kamisah
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
39
|
Mostafa DG, Satti HH. Resolvin D1 Prevents the Impairment in the Retention Memory and Hippocampal Damage in Rats Fed a Corn Oil-Based High Fat Diet by Upregulation of Nrf2 and Downregulation and Inactivation of p 66Shc. Neurochem Res 2020; 45:1576-1591. [PMID: 32253649 DOI: 10.1007/s11064-020-03022-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/27/2020] [Accepted: 04/02/2020] [Indexed: 12/31/2022]
Abstract
This study investigated the effect of a high-fat diet rich in corn oil (CO-HFD) on the memory retention and hippocampal oxidative stress, inflammation, and apoptosis in rats, and examined if the underlying mechanisms involve modulating Resolvin D1 (RvD1) levels and activation of p66Shc. Also, we tested if co-administration of RvD1 could prevent these neural adverse effects induced by CO-HFD. Adult male Wistar rats were divided into 4 groups (n = 18/each) as control fed standard diet (STD) (3.82 kcal/g), STD + RvD1 (0.2 µg/Kg, i.p/twice/week), CO-HFD (5.4 kcal/g), and CO-HFD + RvD1. All treatments were conducted for 8 weeks. With normal fasting glucose levels, CO-HFD induced hyperlipidemia, hyperinsulinemia, increased HOMA-IRI and reduced the rats' memory retention. In parallel, CO-HFD increased levels of reactive oxygen species (ROS), malondialdehyde (MDA), cytoplasmic cytochrome-c, and cleaved caspase-3 and significantly decreased levels of glutathione (GSH), Bcl-2, and manganese superoxide dismutase (MnSOD) in rats' hippocampi. Besides, CO-HFD significantly reduced hippocampal levels of docosahexaenoic acid (DHA) and RvD1, as well as total protein levels of Nrf2 and significantly increased nuclear protein levels of p-NF-κB. Concomitantly, CO-HFD increased hippocampal protein levels of p-JNK, p53, p66Shc, p-p66Shc, and NADPH oxidase. However, without altering plasma and serum levels of glucose, insulin, and lipids, co-administration of RvD1 to CO-HFD completely reversed all these events. It also resulted in similar effects in the STD fed-rats. In conclusion, CO-HFD impairs memory function and induces hippocampal damage by reducing levels of RvD1 and activation of JNK/p53/p66Shc/NADPH oxidase, effects that are prevented by co-administration of RvD1.
Collapse
Affiliation(s)
- Dalia G Mostafa
- Department of Medical Physiology, College of Medicine, Kingdom of Saudi Arabia, King Khalid University, P.O. Box 3340, Abha, 61421, Kingdom of Saudi Arabia. .,Department of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, Egypt.
| | - Huda H Satti
- Department of Pathology, College of Medicine, Kingdom of Saudi Arabia, King Khalid University, P.O.Box 3340, Abha, 61421, Kingdom of Saudi Arabia.,Department of Pathology, Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| |
Collapse
|