1
|
Mukherjee AG, Mishra S, Gopalakrishnan AV, Kannampuzha S, Murali R, Wanjari UR, B S, Vellingiri B, Madhyastha H, Kanagavel D, Vijayan M. Unraveling the mystery of citrate transporters in Alzheimer's disease: An updated review. Ageing Res Rev 2025; 107:102726. [PMID: 40073978 DOI: 10.1016/j.arr.2025.102726] [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: 06/25/2024] [Revised: 12/26/2024] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
A key molecule in cellular metabolism, citrate is essential for lipid biosynthesis, energy production, and epigenetic control. The etiology of Alzheimer's disease (AD), a progressive neurodegenerative illness marked by memory loss and cognitive decline, may be linked to dysregulated citrate transport, according to recent research. Citrate transporters, which help citrate flow both inside and outside of cells, are becoming more and more recognized as possible participants in the molecular processes underlying AD. Citrate synthase (CS), a key enzyme in the tricarboxylic acid (TCA) cycle, supports mitochondrial function and neurotransmitter synthesis, particularly acetylcholine (ACh), essential for cognition. Changes in CS activity affect citrate availability, influencing energy metabolism and neurotransmitter production. Choline, a precursor for ACh, is crucial for neuronal function. Lipid metabolism, oxidative stress reactions, and mitochondrial function can all be affected by aberrant citrate transport, and these changes are linked to dementia. Furthermore, the two main pathogenic characteristics of AD, tau hyperphosphorylation and amyloid-beta (Aβ) aggregation, may be impacted by disturbances in citrate homeostasis. The goal of this review is to clarify the complex function of citrate transporters in AD and provide insight into how they contribute to the development and course of the illness. We aim to provide an in-depth idea of which particular transporters are dysregulated in AD and clarify the functional implications of these dysregulated transporters in brain cells. To reduce neurodegenerative processes and restore metabolic equilibrium, we have also discussed the therapeutic potential of regulating citrate transport. Gaining insight into the relationship between citrate transporters and the pathogenesis of AD may help identify new indicators for early detection and creative targets for treatment. This study offers hope for more potent ways to fight this debilitating illness and is a crucial step in understanding the metabolic foundations of AD.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Shatakshi Mishra
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, VIT, Vellore 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| | - Sandra Kannampuzha
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Reshma Murali
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Stany B
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, VIT, Vellore 632014, India
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda, Punjab 151401, India
| | - Harishkumar Madhyastha
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki 8891692, Japan
| | - Deepankumar Kanagavel
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, VIT, Vellore 632014, India
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
2
|
Coyotl-Pérez WA, Ángeles-López YI, Luna-Suárez S, Rosas-Cárdenas FDF, Villa-Ruano N. Volatilomics of Capsicum pubescens Plants Infested by Solenopsis geminata: Unraveling the Role of Oleic and Palmitic Acids in Plant-Fire Ant Interaction. Chem Biodivers 2025; 22:e202402380. [PMID: 39665862 DOI: 10.1002/cbdv.202402380] [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: 09/21/2024] [Revised: 11/21/2024] [Accepted: 12/12/2024] [Indexed: 12/13/2024]
Abstract
Solenopsis geminata is an aggressive pest of manzano pepper (Capsicum pubescens) crops. Herein, we report on the volatilomics profiling of manzano pepper plants obtained during S. geminata infestation by solid-phase microextraction coupled with gas chromatography-mass spectrometry. As a result, 68 volatile organic compounds were identified from ants, non-infested plants, and infested plants, including terpenes, esters, steroids, aldehydes, phenylpropanoids, and fatty acids. As a remarkable finding, oleic and palmitic acids were the main compounds released during ant infestation. These fatty acids were evaluated as biocidal or repellent agents under in vitro and in situ conditions. From these experiments, the biocidal effect of palmitic acid was more potent (median lethal dose [LC50], 0.97 mg/cm2) than that of oleic acid (LC50, 5.03 mg/cm2) on S. geminata workers. Nevertheless, only oleic acid had a repellent effect under in situ conditions (p < 0.01). Our results represent new insights into the role of both fatty acids in manzano pepper defense mechanisms.
Collapse
Affiliation(s)
- Wendy Abril Coyotl-Pérez
- Centro de Investigación en Biotecnología Aplicada, Instituto Politécnico Nacional (IPN), Ex-Hacienda San Juan Molino Carretera Estatal, Santa Inés Tecuexcomac, Tepetitla, Tlaxcala, Mexico
| | - Yesenia Ithaí Ángeles-López
- Dirección de Innovación y Transferencia de Conocimiento, Benemérita Universidad Autónoma de Puebla (BUAP), Prolongación 24 sur y Av. San Claudio, Puebla, Mexico
| | - Silvia Luna-Suárez
- Centro de Investigación en Biotecnología Aplicada, Instituto Politécnico Nacional (IPN), Ex-Hacienda San Juan Molino Carretera Estatal, Santa Inés Tecuexcomac, Tepetitla, Tlaxcala, Mexico
| | - Flor de Fátima Rosas-Cárdenas
- Centro de Investigación en Biotecnología Aplicada, Instituto Politécnico Nacional (IPN), Ex-Hacienda San Juan Molino Carretera Estatal, Santa Inés Tecuexcomac, Tepetitla, Tlaxcala, Mexico
| | - Nemesio Villa-Ruano
- CONAHCyT-Dirección de Innovación y Transferencia de Conocimiento, Benemérita Universidad Autónoma, de Puebla, Prolongación de la 24 Sur y Av. San Claudio, Puebla, Mexico
| |
Collapse
|
3
|
Wang D, Irewole EA, Bays LD, Smith MD, Schreurs BG. A long-term mild high-fat diet facilitates rabbit discrimination learning and alters glycerophospholipid metabolism. Neurobiol Learn Mem 2025; 219:108053. [PMID: 40228735 DOI: 10.1016/j.nlm.2025.108053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/28/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Previous reports have shown an association between a Western high-fat diet (HFD) and poor cognitive performance. So far, there are no reports of whether a mild HFD can affect rabbit learning and hippocampal metabolic profile. This study was designed to explore whether feeding a mild HFD (5 % lard and 5 % soy oil) for 20 weeks affected eyeblink discrimination and discrimination reversal learning and hippocampal metabolic profiles. After 20 weeks on the HFD or a normal control diet, all rabbits received one day of adaptation, 20 daily sessions of two-tone discrimination (1-kHz CS + followed by air puff and 8-kHz CS- not followed by air puff), a rest day, and then 40 daily sessions of discrimination reversal (8-kHz CS + and 1-kHz CS-). Compared to rabbits fed a regular chow diet, rabbits fed a mild HFD showed better discrimination evidenced by higher responding to CS+, lower responding to CS-, and a larger discrimination index (CS+ - CS-). Widely targeted metabolomics analysis identified 1805 metabolites in the hippocampus, and significant HFD-induced changes in 162 and 165 differential metabolites in males and females, respectively. These included glycerophospholipids and fatty acyls. KEGG enrichment analysis showed glycerophospholipid metabolism (ko00564) was significantly enriched in the HFD group notably lysophosphatidylethanolamine and lysophosphatidylcholine. In summary, our data show a long-term mild HFD facilitated discrimination learning in rabbits without inducing a metabolic syndrome, and altered the hippocampal metabolic profile, which may affect neuronal cell membrane lipids and behavioral performance.
Collapse
Affiliation(s)
- Desheng Wang
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA.
| | - Ezekiel A Irewole
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Logan D Bays
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - MacKinzie D Smith
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Bernard G Schreurs
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
4
|
Vallazhath A, Thimmappa PY, Joshi HB, Hebbar KR, Nayak A, Umakanth S, Saoji AA, Manjunath NK, Hadapad BS, Joshi MB. A comprehensive review on the implications of Yogic/Sattvic diet in reducing inflammation in type 2 diabetes. Nutr Diabetes 2025; 15:14. [PMID: 40216734 PMCID: PMC11992243 DOI: 10.1038/s41387-025-00371-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
Chronic inflammation in type 2 diabetes (T2D), characterized by constitutively activated immune cells and elevated pro-inflammatory mediators along with hyperglycaemia and increased free fatty acids and branched chain amino acid levels, significantly alters the immuno-metabolic axis. Over the years, dietary intervention has been explored as an effective strategy for managing T2D. Evidence from experimental and clinical studies indicates that various diets, including Mediterranean, Nordic, Palaeolithic and ketogenic diets, increase insulin sensitivity, decrease gluconeogenesis, and adiposity, and exert anti-inflammatory effects, thus preserving immuno-metabolic homeostasis in individuals with T2D. Indian dietary sources are categorized as Sattvic, Rajasic, and Tamasic, depending on their impact on health and behaviour. The Yogic diet, commonly recommended during yoga practice, is predominantly Sattvic, emphasizing plant-based whole foods while limiting processed and high-glycaemic-index items. Yogic diet is also recommended for Mitahara, emphasizing mindful eating, which is attributed to calorie restriction. Adopting a Yogic diet, featuring low-fat vegetarian principles, strongly reduces inflammatory mediator levels. This diet not only ameliorates insulin resistance and maintains a healthy body weight but also regulates immunomodulation, enhances gut microbiome diversity and provides essential phytonutrients, collectively preventing inflammation. Although, preliminary studies show aforementioned beneficial role of Yogic diet in improving diabetes associated metabolic and inflammatory changes, precise cellular and molecular mechanisms are not yet understood. Hence, further studies are warranted to decipher the mechanisms. This review summarizes the multiple roles of Yogic diet and related dietary components in mitigating inflammation and enhancing glycaemic control in T2D.
Collapse
Affiliation(s)
- Anupama Vallazhath
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Pooja Yedehalli Thimmappa
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Harshit B Joshi
- Division of Ayurveda, Centre for Integrative Medicine and Research, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Krishna Raghava Hebbar
- Division of Ayurveda, Centre for Integrative Medicine and Research, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Anupama Nayak
- Division of Ayurveda, Centre for Integrative Medicine and Research, Manipal Academy of Higher Education, Manipal, 576104, India
| | | | - Apar Avinash Saoji
- Swami Vivekananda Yoga Anusandhana Samsthana, Bangalore, 560105, Karnataka, India
| | | | - Basavaraj S Hadapad
- Division of Ayurveda, Centre for Integrative Medicine and Research, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Manjunath B Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
- Centre for Ayurveda Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
| |
Collapse
|
5
|
Miyamoto J, Ando Y, Yamano M, Nishida A, Murakami K, Kimura I. Acidipropionibacterium acidipropionici, a propionate-producing bacterium, contributes to GPR41 signaling and metabolic regulation in high-fat diet-induced obesity in mice. Front Nutr 2025; 12:1542196. [PMID: 40248033 PMCID: PMC12003125 DOI: 10.3389/fnut.2025.1542196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/19/2025] [Indexed: 04/19/2025] Open
Abstract
Obesity is a major healthcare problem worldwide and is induced by excess energy intake, resulting in gut microbial composition and microbial diversity changes. Through fermentation of dietary fibers, short-chain fatty acids (SCFAs) act as host energy sources and signaling molecules via G protein-coupled receptors such as GPR41. Acidipropionibacterium acidipropionici is widely used in many applications; however, in vivo studies on the beneficial effect of A. acidipropionici via propionate production and host energy homeostasis are unclear. Therefore, this study aimed to investigate the beneficial metabolic effects of A. acidipropionici by focusing on GPR41 signaling in a high-fat diet (HFD)-induced obesity mouse model. Here, we demonstrated that A. acidipropionici OB7439 improved host metabolism in HFD-induced obesity in mice. The intake of A. acidipropionici OB7439 improved metabolism in HFD-induced obese mice by increasing propionate production, regulating glucose tolerance, and inhibiting hepatic inflammation via GPR41 signaling. Our findings shed light on the potential of using A. acidipropionici OB7439 as an SCFA producer for the prevention and treatment of metabolic disorders. Based on these results, we suggest that A. acidipropionici may be a potential therapeutic bacterium that inhibits obesity and modulates the gut microbial community.
Collapse
Affiliation(s)
- Junki Miyamoto
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan
| | - Yuna Ando
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Mayu Yamano
- Department of Molecular Endocrinology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Akari Nishida
- Department of Molecular Endocrinology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Kota Murakami
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan
| | - Ikuo Kimura
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Department of Molecular Endocrinology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
6
|
Braden-Kuhle PN, Lacy VA, Brice KN, Bertrand ME, Uras HB, Shoffner C, Fischer BE, Rana A, Willis JL, Boehm GW, Chumley MJ. A Mediterranean-style diet protects against cognitive and behavioral deficits, adiposity, and Alzheimer's disease-related markers, compared to a macronutrient-matched typical American diet in C57BL/6J mice. J Alzheimers Dis 2025; 104:678-697. [PMID: 40007076 DOI: 10.1177/13872877251319467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
BackgroundResearch suggests that modifying risk factors may prevent or delay up to 40% of dementia cases, including Alzheimer's disease (AD). Thus, understanding the potential of healthful dietary patterns, like the Mediterranean diet (MD), in AD prevention is crucial. While supplementation of individual Mediterranean foods has demonstrated efficacy in reducing AD biomarkers and cognitive impairment in rodents, the effects of a comprehensive MD warrant further investigation. Additionally, while rodent studies often use a "Western diet" as a model for the typical American diet (TAD), these diets generally exceed the macronutrient densities of typical American consumption, particularly in fats and carbohydrates.ObjectiveTo better reflect human diets, we developed two diets for mice that more closely mirrored the macronutrient composition of the traditional MD or the TAD, each with matched macronutrient profiles (50% kcal from carbohydrates, 35% kcal from fat, 15% kcal from protein), and distinct food sources from Mediterranean regions or the U.S., respectively.MethodsMale C57BL/6J mice were randomly assigned to one diet (MD or TAD) at weaning (21 days of age), which they consumed for six months.ResultsCompared to the TAD, MD animals had lower body weight, abdominal and hepatic fat, serum TNF-α, and central Aβ1-42, while also exhibiting enhanced exploratory behavior, reduced anxiety-like behavior, and preserved spatial memory. The MD also protected against LPS-induced central inflammation and BDNF loss.ConclusionsThese findings suggest that a comprehensive MD provides protection against metabolic and AD-related markers in wildtype mice, despite matched caloric availability to the TAD.
Collapse
Affiliation(s)
- Paige N Braden-Kuhle
- Department of Psychology, Vanguard University of Southern California, Costa Mesa, CA, USA
| | - Vivienne A Lacy
- Department of Biology, Texas Christian University, Fort Worth, TX, USA
| | - Kelly N Brice
- Department of Psychological Sciences, Rice University, Houston, TX, USA
| | - Morgan E Bertrand
- Department of Biology, Texas Christian University, Fort Worth, TX, USA
| | - Hatice Buse Uras
- Department of Psychology, Texas Christian University, Fort Worth, TX, USA
| | - Catherine Shoffner
- Department of Psychology, Texas Christian University, Fort Worth, TX, USA
| | | | - Ashish Rana
- Department of Psychology, Texas Christian University, Fort Worth, TX, USA
| | - Jada L Willis
- Department of Nutritional Sciences, Texas Christian University, Fort Worth, TX, USA
| | - Gary W Boehm
- Department of Psychology, Texas Christian University, Fort Worth, TX, USA
| | - Michael J Chumley
- Department of Biology, Texas Christian University, Fort Worth, TX, USA
| |
Collapse
|
7
|
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
|
8
|
Dreux V, Lefebvre C, Breemeersch CE, Salaün C, Bôle-Feysot C, Guérin C, Déchelotte P, Goichon A, Coëffier M, Langlois L. Sex-dependent effects of a high-fat diet on the hypothalamic response in mice. Biol Sex Differ 2025; 16:17. [PMID: 40001261 PMCID: PMC11854408 DOI: 10.1186/s13293-025-00699-3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Sex differences in rodent models of diet-induced obesity are still poorly documented, particularly regarding how central mechanisms vary between sexes in response to an obesogenic diet. Here, we wanted to determine whether obese phenotype and hypothalamic response to a high-fat diet (HFD) differed between male and female C57Bl/6J mice. Mice were exposed to either a 60% HFD or a standard diet first for both a long- (14 weeks) and shorter-periods of time (3, 7, 14 and 28 days). Analysis of the expression profile of key neuronal, glial and inflammatory hypothalamic markers was performed using RT-qPCR. In addition, astrocytic and microglial morphology was examined in the arcuate nucleus. Monitoring of body weight and composition revealed that body weight and fat mass gain appeared earlier and was more pronounced in male mice. After 14 weeks of HFD exposure, normalized increase of body weight reached similar levels between male and female mice. Overall, both sexes under HFD displayed a decrease of orexigenic neuropeptides expression while an increase in Pomc gene expression was observed only in female mice. In addition, changes in the expression of hypothalamic inflammatory markers were relatively modest. We also reported that the glial cell markers expression and morphology were affected by HFD in a sex- and time dependent manner, suggesting a more pronounced glial cell activation in female mice. Taken together, these data show that male and female mice responded differently to HFD exposure, both on short- and long-term and suggest that a strong inflammatory hypothalamic profile is not systematically present in diet-induced obesity models. Nevertheless, in addition to these present data, the underlying mechanisms should be deciphered in further investigations.
Collapse
Affiliation(s)
- Virginie Dreux
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis, F-76000, Rouen, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), F-76000, Rouen, France
| | - Candice Lefebvre
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis, F-76000, Rouen, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), F-76000, Rouen, France
| | - Charles-Edward Breemeersch
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis, F-76000, Rouen, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), F-76000, Rouen, France
| | - Colin Salaün
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis, F-76000, Rouen, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), F-76000, Rouen, France
| | - Christine Bôle-Feysot
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis, F-76000, Rouen, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), F-76000, Rouen, France
| | - Charlène Guérin
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis, F-76000, Rouen, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), F-76000, Rouen, France
| | - Pierre Déchelotte
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis, F-76000, Rouen, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), F-76000, Rouen, France
- Department of Nutrition, CHU Rouen, F-76000, Rouen, France
| | - Alexis Goichon
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis, F-76000, Rouen, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), F-76000, Rouen, France
| | - Moïse Coëffier
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis, F-76000, Rouen, France
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), F-76000, Rouen, France
- Department of Nutrition, CHU Rouen, F-76000, Rouen, France
| | - Ludovic Langlois
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis, F-76000, Rouen, France.
- Univ Rouen Normandie, Institute for Research and Innovation in Biomedicine (IRIB), F-76000, Rouen, France.
| |
Collapse
|
9
|
Crowder SL, Gudenkauf LM, Hoogland AI, Han HS, Small BJ, Carson TL, Parker NH, Booth-Jones M, Jim HSL. Cancer-Related Cognitive Impairment and the Potential of Dietary Interventions for the Prevention and Mitigation of Neurodegeneration. Cancer Res 2025; 85:203-217. [PMID: 39570793 DOI: 10.1158/0008-5472.can-24-3041] [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: 08/21/2024] [Revised: 10/02/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025]
Abstract
Approximately 35% of long-term cancer survivors experience ongoing cancer-related cognitive impairment (CRCI). Yet, few efficacious interventions exist to prevent or ameliorate CRCI. The underlying biological processes driving CRCI are complex and are reported to include changes in brain structure and function, increased oxidative stress and inflammation, and alterations in gut microbiome composition. Some of the mechanisms promoting CRCI have the potential to be modified through behavioral changes, such as dietary changes. Compelling evidence from randomized controlled trials and observational research supports the positive impacts of the Mediterranean-DASH Intervention for Neurodegenerative Delay diet on cognition outside of the context of cancer, but studies investigating the Mediterranean-DASH Intervention for Neurodegenerative Delay diet as an intervention for people who experience CRCI are lacking. This review examines the current state of the science for cognitive outcomes of dietary interventions in aging populations and discusses future opportunities to adapt these interventions to cancer populations.
Collapse
Affiliation(s)
- Sylvia L Crowder
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Lisa M Gudenkauf
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Aasha I Hoogland
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Hyo S Han
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Brent J Small
- School of Nursing, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina
| | - Tiffany L Carson
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Nathan H Parker
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Margaret Booth-Jones
- Department of Supportive Care Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Heather S L Jim
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
10
|
Wong GC, Bearzatto B, Gala JL, Delzenne NM, Van Hul M, Cani PD. Obesity phenotype and gut microbiota alterations are not associated with anxiety-like behaviour in high-fat diet-fed mice. Food Funct 2025; 16:268-281. [PMID: 39655876 DOI: 10.1039/d4fo04461d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Anxiety is a common co-morbidity with obesity and metabolic disease, and can lead to a significant impact on quality of life. The vast differences in the gut microbiota between obese and control individuals provide a potential avenue for therapeutic intervention. A high-fat diet (HFD) in rodent models have been shown to induce anxiety-like behaviour and has been tested through an array of distinct behavioural tests such as the elevated plus maze test, light-dark test and open field test. Despite differences in testing and assessment parameters, the behavioural outcomes have previously yielded similar results. Recent evidence suggests that HFD has an anxiolytic effect on mice, complicating the model. Here, we aimed to confirm whether HFD-fed mice are more susceptible to presenting anxiety-like behaviours. Our findings showed no significant differences in behaviour, plasma corticosterone and inflammation markers between HFD and control diet (CTD) mice, despite considerable differences in adiposity and faecal microbial communities. Additionally, daily oral gavage is one of the most common methods for testing bacterial probiotics in rodent models, but this handling could potentially also cause stress to the mice. Thus, we investigated if daily oral gavage could mask differences in HFD and CTD mice. We found no significant differences in weight, fat mass or anxiety-like behaviour in CTD-fed mice with or without daily oral gavage.
Collapse
Affiliation(s)
- Giselle C Wong
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), 1200 Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Bertrand Bearzatto
- UCLouvain, Université catholique de Louvain, Institute of Experimental and Clinical Research (IREC), Centre des Technologies Moléculaires Appliquées (CTMA), 1200 Brussels, Belgium
| | - Jean-Luc Gala
- UCLouvain, Université catholique de Louvain, Institute of Experimental and Clinical Research (IREC), Centre des Technologies Moléculaires Appliquées (CTMA), 1200 Brussels, Belgium
| | - Nathalie M Delzenne
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), 1200 Brussels, Belgium.
| | - Matthias Van Hul
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), 1200 Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Patrice D Cani
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), 1200 Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO Department, WEL Research Institute, Wavre, Belgium
- UCLouvain, Université catholique de Louvain, Institute of Experimental and Clinical Research (IREC), 1200 Brussels, Belgium
| |
Collapse
|
11
|
Borrego-Ruiz A, Borrego JJ. Human gut microbiome, diet, and mental disorders. Int Microbiol 2025; 28:1-15. [PMID: 38561477 PMCID: PMC11775079 DOI: 10.1007/s10123-024-00518-6] [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: 02/03/2024] [Revised: 03/15/2024] [Accepted: 03/22/2024] [Indexed: 04/04/2024]
Abstract
Diet is one of the most important external factor shaping the composition and metabolic activities of the gut microbiome. The gut microbiome plays a crucial role in host health, including immune system development, nutrients metabolism, and the synthesis of bioactive molecules. In addition, the gut microbiome has been described as critical for the development of several mental disorders. Nutritional psychiatry is an emerging field of research that may provide a link between diet, microbial function, and brain health. In this study, we have reviewed the influence of different diet types, such as Western, Mediterranean, vegetarian, and ketogenic, on the gut microbiota composition and function, and their implication in various neuropsychiatric and psychological disorders.
Collapse
Affiliation(s)
- Alejandro Borrego-Ruiz
- Departamento de Psicología Social y de las Organizaciones, Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
| | - Juan J Borrego
- Departamento de Microbiología, Universidad de Málaga. Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina BIONAND, Málaga, Spain.
| |
Collapse
|
12
|
Kilic EB, Koksal E. The Interaction Between Attention Deficit and Hyperactivity Disorder and Nutrition. Curr Nutr Rep 2024; 14:1. [PMID: 39508912 DOI: 10.1007/s13668-024-00592-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
PURPOSE OF REVIEW This review aims to explore the relationship between Attention Deficit Hyperactivity Disorder (ADHD) and nutrition. ADHD, a neurodevelopmental disorder, has been examined in relation to dietary factors through various metabolic pathways, with a focus on the role of nutrition in symptom management. Unhealthy dietary patterns, particularly those characteristics of Western diets, are believed to exacerbate ADHD symptoms through these mechanisms. In contrast, dietary interventions such as intermittent fasting, which offer greater flexibility in application, have been proposed as potential strategies to alleviate ADHD symptoms. While further research in this area is expected to contribute significantly to the field, this review also provides researchers with a brief perspective on the challenges and limitations associated with experimental ADHD studies. Therefore, this study aims to offer a comprehensive evaluation of the interaction between ADHD and nutrition, providing researchers with an integrative approach to the topic. RECENT FINDINGS Western dietary patterns have been found to negatively impact gut barrier integrity, synaptic plasticity, insulin resistance, and oxidative stress. On the other hand, the intermittent fasting diet model, which offers practical flexibility, is thought to be a potentially supportive treatment in managing ADHD. Furthermore, it has been concluded that various experimental models are available for ADHD research, and researchers must work within these limitations. Western diets, particularly in their negative impact on synaptic plasticity and other key metabolic pathways involved in ADHD, can worsen the disorder's symptoms. Intermittent fasting emerges as a promising dietary alternative that may mitigate these adverse effects.
Collapse
Affiliation(s)
- Enes Bahadir Kilic
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gaziantep University, Gaziantep, 27310, Turkey.
| | - Eda Koksal
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara, 06490, Turkey
| |
Collapse
|
13
|
González-Velázquez G, Aguirre-Garrido JF, Oros-Pantoja R, Salinas-Velarde ID, Contreras I, Estrada JA, Soto-Piña AE. Supplementation with inulin reverses cognitive flexibility alterations and modulates the gut microbiota in high-fat-fed mice. Front Behav Neurosci 2024; 18:1445154. [PMID: 39568732 PMCID: PMC11577567 DOI: 10.3389/fnbeh.2024.1445154] [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: 06/06/2024] [Accepted: 10/16/2024] [Indexed: 11/22/2024] Open
Abstract
Introduction Alterations in cognitive performance are associated with inadequate nutritional states and diet composition. Prebiotics, such as inulin, are substances that can modulate the gut microbiome and, consequently, brain function by producing metabolites such as short-chain fatty acids (SCFAs). This study aimed to evaluate the effect of supplementation with inulin on cognitive flexibility, body composition, and gut microbiota in a murine model exposed to a high-fat (HF) diet. Methods CD1 mice were divided into five groups: control fed a standard diet (C), high-fat diet (HF), inulin (I), high-fat diet with inulin (HFI), and manipulation control (M). Dietary supplementation was administered for 6 weeks. Cognitive flexibility was assessed using the Attentional Set-Shifting Test (AST). In addition, body composition was measured via electrical bioimpedance and adipose tissue compartments of each mouse were removed and weighed. Finally, gut microbiota metataxonomic was analyzed through metataxonomic bacterial 16S rRNA sequencing. Results We observed that HF group required more AST trials than the C, HFI, and I groups in the compound discrimination (CD) and extra-dimensional (ED) stages. Notably, the HFI group required fewer trials than the HF group in the ED stage (p = 0.0187). No significant differences in overall body composition were observed between the groups. However, the percentage of gonadal and peritoneal adipose tissue was significantly higher in the HF and I groups compared to the C group. Statistically significant differences in alpha diversity for gut microbiota were observed using the Shannon, Simpson, and Chao1 indices. The I group showed a decrease in bacterial diversity compared to the HF group. While no differences were observed between groups in the phyla Bacillota and Bacteroidotes, Clostridium bacteria represented a lower proportion of sequences in the I group compared to the C group. Additionally, Lactobacillus represented a lower proportion of sequences in the HF group compared to the C and I groups. Discussion These findings suggest that supplementation with inulin could be a useful approach to mitigate the negative effects of an HF diet on cognitive flexibility and modulate gut microbiota composition.
Collapse
Affiliation(s)
| | - José Félix Aguirre-Garrido
- Departamento de Ciencias Ambientales, Universidad Autónoma Metropolitana-Lerma, Lerma, Estado de México, Mexico
| | - Rigoberto Oros-Pantoja
- Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico
| | | | - Irazú Contreras
- Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico
| | - José Antonio Estrada
- Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico
| | | |
Collapse
|
14
|
O'Dowd A, O'Connor DMA, Hirst RJ, Setti A, Kenny RA, Newell FN. Nutrition is associated with differences in multisensory integration in healthy older adults. Nutr Neurosci 2024; 27:1226-1236. [PMID: 38386286 DOI: 10.1080/1028415x.2024.2316446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Diet can influence cognitive functioning in older adults and is a modifiable risk factor for cognitive decline. However, it is unknown if an association exists between diet and lower-level processes in the brain underpinning cognition, such as multisensory integration. We investigated whether temporal multisensory integration is associated with daily intake of fruit and vegetables (FV) or products high in fat/sugar/salt (FSS) in a large sample (N = 2,693) of older adults (mean age = 64.06 years, SD = 7.60; 56% female) from The Irish Longitudinal Study on Ageing (TILDA). Older adults completed a Food Frequency Questionnaire from which the total number of daily servings of FV and FSS items respectively was calculated. Older adults' susceptibility to the Sound Induced Flash Illusion (SIFI) measured the temporal precision of audio-visual integration, which included three audio-visual Stimulus Onset Asynchronies (SOAs): 70, 150 and 230 ms. Older adults who self-reported a higher daily consumption of FV were less susceptible to the SIFI at the longest versus shortest SOAs (i.e. increased temporal precision) compared to those reporting the lowest daily consumption (p = .013). In contrast, older adults reporting a higher daily consumption of FSS items were more susceptible to the SIFI at the longer versus shortest SOAs (i.e. reduced temporal precision) compared to those reporting the lowest daily consumption (p < .001). The temporal precision of multisensory integration is differentially associated with levels of daily consumption of FV versus products high in FSS, consistent with broader evidence that habitual diet is associated with brain health.
Collapse
Affiliation(s)
- Alan O'Dowd
- School of Psychology and Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, Dublin, Ireland
| | - Deirdre M A O'Connor
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, Dublin, Ireland
- Mercer Institute for Successful Ageing, St. James Hospital, Dublin, Ireland
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Rebecca J Hirst
- School of Psychology and Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, Dublin, Ireland
| | - Annalisa Setti
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, Dublin, Ireland
- School of Applied Psychology, University College Cork, Cork, Ireland
| | - Rose Anne Kenny
- The Irish Longitudinal Study on Ageing, Trinity College Dublin, Dublin, Ireland
- Mercer Institute for Successful Ageing, St. James Hospital, Dublin, Ireland
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Fiona N Newell
- School of Psychology and Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
15
|
Andraka JM, Sharma N, Marchalant Y. New Insights on the Effects of Krill Oil Supplementation, a High-Fat Diet, and Aging on Hippocampal-Dependent Memory, Neuroinflammation, Synaptic Density, and Neurogenesis. Int J Mol Sci 2024; 25:11554. [PMID: 39519107 PMCID: PMC11545834 DOI: 10.3390/ijms252111554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Krill oil (KO) has been described as having the potential to ameliorate the detrimental consequences of a high-fat diet (HFD) on the aging brain, though the magnitude and mechanism of this benefit is unclear. We thus hypothesized that dietary KO supplementation could counteract the effects of cognitive aging and an HFD on spatial learning, neuroinflammation, neurogenesis, and synaptic density in the cortex and hippocampus of aged rats. Sixteen-month-old Sprague Dawley rats were fed for 12 weeks while being divided into four groups: control (CON); control with KO supplementation (CONKO); high-fat diet (HF); and high-fat diet with KO supplementation (HFKO). We measured food consumption, body mass, spatial memory (Morris water maze), microglia, neurogenesis, cytokine concentrations, and synaptic markers (post-synaptic density-95 and synaptophysin). Predictably, an HFD did induce significant differences in body weights, with the high-fat groups gaining more weight than the low-fat controls. However, KO supplementation did not produce significant changes in the other quantified parameters. Our results demonstrate that the dietary KO dose provided in the current study does not benefit hippocampal or cortical functions in an aging model. Our results provide a benchmark for future dosing protocols that may eventually prove to be beneficial.
Collapse
Affiliation(s)
- John M. Andraka
- Department of Physical Therapy, Central Michigan University, Mt. Pleasant, MI 48859, USA
- Neuroscience Program, Central Michigan University, Mt. Pleasant, MI 48859, USA; (N.S.); (Y.M.)
| | - Naveen Sharma
- Neuroscience Program, Central Michigan University, Mt. Pleasant, MI 48859, USA; (N.S.); (Y.M.)
- School of Health Sciences, Central Michigan University, Mt. Pleasant, MI 48859, USA
| | - Yannick Marchalant
- Neuroscience Program, Central Michigan University, Mt. Pleasant, MI 48859, USA; (N.S.); (Y.M.)
- Psychology Department, Central Michigan University, Mt. Pleasant, MI 48859, USA
| |
Collapse
|
16
|
Llanquinao J, Jara C, Cortés-Díaz D, Kerr B, Tapia-Rojas C. Contrasting Effects of an Atherogenic Diet and High-Protein/Unsaturated Fatty Acids Diet on the Accelerated Aging Mouse Model SAMP8 Phenotype. Neurol Int 2024; 16:1066-1085. [PMID: 39452682 PMCID: PMC11510401 DOI: 10.3390/neurolint16050080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 10/26/2024] Open
Abstract
Background/Objectives: Aging has been extensively studied, with a growing interest in memory impairment by a neurobiological approach. Mitochondrial dysfunction is a hallmark of aging, contributing to the aging phenotype; therefore, mitochondrial interventions seem fundamental. The diet is a physiological approximation for modifying mitochondria, which could impact the age-related phenotype. Methods: We studied two diets with low-carbohydrate and high-fat compositions, differing in the amount of protein and the fat type disposable-the atherogenic diet Cocoa (high protein/high saturated fat/high cholesterol) and the South Beach diet (very high-protein/high-unsaturated fat)-on oxidative stress, mitochondrial state, and hippocampus-dependent memory in 3-month-old Senescence-Accelerated Mouse Model (SAMP8) seed over 3 months to determine their pro- or anti-aging effects. Results: Despite its bad reputation, the Cocoa diet reduces the reactive oxygen species (ROS) content without impacting the energy state and hippocampus-dependent spatial acuity. In contrast to the beneficial impact proposed for the South Beach diet, it induced a pro-aging phenotype, increasing oxidative damage and the levels of NR2B subunit of the NMDA, impairing energy and spatial acuity. Surprisingly, despite the negative changes observed with both diets, this led to subtle memory impairment, suggesting the activation of compensatory mechanisms preventing more severe cognitive decline. Conclusions: Our results demonstrated that diets usually considered good could be detrimental to the onset of aging. Also, probably due to the brain plasticity of non-aged animals, they compensate for the damage, preventing a more aggravated phenotype. Nevertheless, these silent changes could predispose or increase the risk of suffering pathologies at advanced age.
Collapse
Affiliation(s)
- Jesús Llanquinao
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia-Santiago 7510157, Chile; (J.L.); (C.J.); (D.C.-D.)
- Laboratory of Neuroendocrinology and Metabolism, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia-Santiago 7510157, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida (FCV), Avenida Del Valle Norte #725, Huechuraba, Santiago 8580702, Chile
| | - Claudia Jara
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia-Santiago 7510157, Chile; (J.L.); (C.J.); (D.C.-D.)
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida (FCV), Avenida Del Valle Norte #725, Huechuraba, Santiago 8580702, Chile
| | - Daniela Cortés-Díaz
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia-Santiago 7510157, Chile; (J.L.); (C.J.); (D.C.-D.)
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida (FCV), Avenida Del Valle Norte #725, Huechuraba, Santiago 8580702, Chile
| | - Bredford Kerr
- Laboratory of Neuroendocrinology and Metabolism, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia-Santiago 7510157, Chile
| | - Cheril Tapia-Rojas
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia-Santiago 7510157, Chile; (J.L.); (C.J.); (D.C.-D.)
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida (FCV), Avenida Del Valle Norte #725, Huechuraba, Santiago 8580702, Chile
| |
Collapse
|
17
|
Ross FC, Mayer DE, Horn J, Cryan JF, Del Rio D, Randolph E, Gill CIR, Gupta A, Ross RP, Stanton C, Mayer EA. Potential of dietary polyphenols for protection from age-related decline and neurodegeneration: a role for gut microbiota? Nutr Neurosci 2024; 27:1058-1076. [PMID: 38287652 DOI: 10.1080/1028415x.2023.2298098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2024]
Abstract
Many epidemiological studies have shown the beneficial effects of a largely plant-based diet, and the strong association between the consumption of a Mediterranean-type diet with healthy aging including a lower risk of cognitive decline. The Mediterranean diet is characterized by a high intake of olive oil, fruits and vegetables and is rich in dietary fiber and polyphenols - both of which have been postulated to act as important mediators of these benefits. Polyphenols are large molecules produced by plants to protect them from environmental threats and injury. When ingested by humans, as little as 5% of these molecules are absorbed in the small intestine with the majority metabolized by the gut microbiota into absorbable simple phenolic compounds. Flavan-3-ols, a type of flavonoid, contained in grapes, berries, pome fruits, tea, and cocoa have been associated with many beneficial effects on several risk factors for cardiovascular disease, cognitive function and brain regions involved in memory formation. Both preclinical and clinical studies suggest that these brain and heart benefits can be attributed to endothelial vascular effects and anti-inflammatory properties among others. More recently the gut microbiota has emerged as a potential modulator of the aging brain and intriguingly polyphenols have been shown to alter microbiota composition and be metabolized by different microbial species. However, there is a need for well controlled studies in large populations to identify predictors of response, particularly given the vast inter-individual variation of human gut microbiota.
Collapse
Affiliation(s)
- F C Ross
- APC Microbiome Ireland, University College Cork, Co. Cork, Ireland
| | - D E Mayer
- Institute of Human Nutrition, Columbia University, New York, USA
| | - J Horn
- Oppenheimer Centre for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, USA
| | - J F Cryan
- APC Microbiome Ireland, University College Cork, Co. Cork, Ireland
- Department Anatomy & Neuroscience, University College Cork, Co. Cork, Ireland
| | - D Del Rio
- Department of Food and Drugs, University of Parma, Parma, Italy
| | - E Randolph
- Oppenheimer Centre for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, USA
| | - C I R Gill
- Nutrition Innovation Centre for Food and Health, Northern Ireland, UK
| | - A Gupta
- Division of Digestive Diseases, UCLA, Los Angeles, USA
- Goodman Luskin Microbiome Center at UCLA, Los Angeles, CA, USA
| | - R P Ross
- APC Microbiome Ireland, University College Cork, Co. Cork, Ireland
| | - C Stanton
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
| | - E A Mayer
- Oppenheimer Centre for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, USA
- Goodman Luskin Microbiome Center at UCLA, Los Angeles, CA, USA
| |
Collapse
|
18
|
Liu X, Tao R, Guo F, Zhang L, Qu J, Li M, Wu X, Wang X, Zhu Y, Wen L, Wang J. Soybean oil induces neuroinflammatory response through brain-gut axis under high-fat diet. J Tradit Complement Med 2024; 14:522-533. [PMID: 39262663 PMCID: PMC11384091 DOI: 10.1016/j.jtcme.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 02/17/2024] [Accepted: 02/28/2024] [Indexed: 09/13/2024] Open
Abstract
Neuroinflammation is considered the principal pathogenic mechanism underlying neurodegenerative diseases, and the incidence of brain disorders is closely linked to dietary fat consumption and intestinal health. To investigate this relationship, 60 8-week-old C57BL/6J mice were subjected to a 20-week dietary intervention, wherein they were fed lard and soybean oil, each at 15% and 35% fat energy. At a dietary fat energy level of 35%, inflammation was observed in both the soybean oil and lard groups. Nevertheless, inflammation was more pronounced in the mice that were administered soybean oil. The process by which nerve cell structure is compromised, inflammatory factors are upregulated, brain antioxidant capacity is diminished, and the TLR4/MyD88/NF-κB p65 inflammatory pathway is activated resulting in damage to the brain-gut barrier. This, in turn, leads to a reduction in the abundance of Akkermansia and unclassified_f_Lachnospiraceae, as well as an increase in Dubosiella abundance, ultimately resulting in brain inflammation and damage. These results suggested that soybean oil induces more severe neuroinflammation compared to lard. Our study demonstrated that, at a dietary fat energy level of 35%, compared to soybean oil, lard could be the healthier option, the outcomes would help provide a reference basis for the selection of residents' daily dietary oil.
Collapse
Affiliation(s)
- Xiangyan Liu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Ran Tao
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Fangrui Guo
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Linyu Zhang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Jianyu Qu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Mengyao Li
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Xiaoran Wu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Xianglin Wang
- Changsha Lvye Biotechnology Co., Ltd., Changsha, 410100, China
| | - Yuanyuan Zhu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Lixin Wen
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Ji Wang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China
| |
Collapse
|
19
|
De Paula GC, Aldana BI, Battistella R, Fernández-Calle R, Bjure A, Lundgaard I, Deierborg T, Duarte JMN. Extracellular vesicles released from microglia after palmitate exposure impact brain function. J Neuroinflammation 2024; 21:173. [PMID: 39014461 PMCID: PMC11253458 DOI: 10.1186/s12974-024-03168-7] [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: 04/29/2024] [Accepted: 07/09/2024] [Indexed: 07/18/2024] Open
Abstract
Dietary patterns that include an excess of foods rich in saturated fat are associated with brain dysfunction. Although microgliosis has been proposed to play a key role in the development of brain dysfunction in diet-induced obesity (DIO), neuroinflammation with cytokine over-expression is not always observed. Thus, mechanisms by which microglia contribute to brain impairment in DIO are uncertain. Using the BV2 cell model, we investigated the gliosis profile of microglia exposed to palmitate (200 µmol/L), a saturated fatty acid abundant in high-fat diet and in the brain of obese individuals. We observed that microglia respond to a 24-hour palmitate exposure with increased proliferation, and with a metabolic network rearrangement that favors energy production from glycolysis rather than oxidative metabolism, despite stimulated mitochondria biogenesis. In addition, while palmitate did not induce increased cytokine expression, it modified the protein cargo of released extracellular vesicles (EVs). When administered intra-cerebroventricularly to mice, EVs secreted from palmitate-exposed microglia in vitro led to memory impairment, depression-like behavior, and glucose intolerance, when compared to mice receiving EVs from vehicle-treated microglia. We conclude that microglia exposed to palmitate can mediate brain dysfunction through the cargo of shed EVs.
Collapse
Affiliation(s)
- Gabriela C De Paula
- Department of Experimental Medical Science (EMV), Faculty of Medicine, Lund University, Sölvegatan 19, BMC C11, Lund, 221 84, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Blanca I Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Roberta Battistella
- Department of Experimental Medical Science (EMV), Faculty of Medicine, Lund University, Sölvegatan 19, BMC C11, Lund, 221 84, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Rosalía Fernández-Calle
- Department of Experimental Medical Science (EMV), Faculty of Medicine, Lund University, Sölvegatan 19, BMC C11, Lund, 221 84, Sweden
| | - Andreas Bjure
- Department of Experimental Medical Science (EMV), Faculty of Medicine, Lund University, Sölvegatan 19, BMC C11, Lund, 221 84, Sweden
| | - Iben Lundgaard
- Department of Experimental Medical Science (EMV), Faculty of Medicine, Lund University, Sölvegatan 19, BMC C11, Lund, 221 84, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Tomas Deierborg
- Department of Experimental Medical Science (EMV), Faculty of Medicine, Lund University, Sölvegatan 19, BMC C11, Lund, 221 84, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - João M N Duarte
- Department of Experimental Medical Science (EMV), Faculty of Medicine, Lund University, Sölvegatan 19, BMC C11, Lund, 221 84, Sweden.
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
20
|
Shetty S, Duesman SJ, Patel S, Huynh P, Toh P, Shroff S, Das A, Chowhan D, Keller B, Alvarez J, Fisher-Foye R, Sebra R, Beaumont K, McAlpine CS, Rajbhandari P, Rajbhandari AK. Sex-specific role of high-fat diet and stress on behavior, energy metabolism, and the ventromedial hypothalamus. Biol Sex Differ 2024; 15:55. [PMID: 39010139 PMCID: PMC11247790 DOI: 10.1186/s13293-024-00628-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 06/11/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Scientific evidence highlights the influence of biological sex on the relationship between stress and metabolic dysfunctions. However, there is limited understanding of how diet and stress concurrently contribute to metabolic dysregulation in both males and females. Our study aimed to investigate the combined effects of high-fat diet (HFD) induced obesity and repeated stress on fear-related behaviors, metabolic, immune, and hypothalamic outcomes in male and female mice. METHODS To investigate this, we used a highly reliable rodent behavioral model that faithfully recapitulates key aspects of post-traumatic stress disorder (PTSD)-like fear. We subjected mice to footshock stressor followed by a weekly singular footshock stressor or no stressor for 14 weeks while on either an HFD or chow diet. At weeks 10 and 14 we conducted glucose tolerance and insulin sensitivity measurements. Additionally, we placed the mice in metabolic chambers to perform indirect calorimetric measurements. Finally, we collected brain and peripheral tissues for cellular analysis. RESULTS We observed that HFD-induced obesity disrupted fear memory extinction, increased glucose intolerance, and affected energy expenditure specifically in male mice. Conversely, female mice on HFD exhibited reduced respiratory exchange ratio (RER), and a significant defect in glucose tolerance only when subjected to repeated stress. Furthermore, the combination of repeated stress and HFD led to sex-specific alterations in proinflammatory markers and hematopoietic stem cells across various peripheral metabolic tissues. Single-nuclei RNA sequencing (snRNAseq) analysis of the ventromedial hypothalamus (VMH) revealed microglial activation in female mice on HFD, while male mice on HFD exhibited astrocytic activation under repeated stress. CONCLUSIONS Overall, our findings provide insights into complex interplay between repeated stress, high-fat diet regimen, and their cumulative effects on health, including their potential contribution to the development of PTSD-like stress and metabolic dysfunctions, emphasizing the need for further research to fully understand these interconnected pathways and their implications for health.
Collapse
Affiliation(s)
- Sanutha Shetty
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Samuel J Duesman
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sanil Patel
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Pacific Huynh
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Pamela Toh
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sanjana Shroff
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anika Das
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Excellence in Youth Education, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Disha Chowhan
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin Keller
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Johana Alvarez
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rachel Fisher-Foye
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Robert Sebra
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristin Beaumont
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cameron S McAlpine
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Prashant Rajbhandari
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Disease Mechanism and Therapeutics Program, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Abha K Rajbhandari
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
21
|
Patel Y, Woo A, Shi S, Ayoub R, Shin J, Botta A, Ketela T, Sung HK, Lerch J, Nieman B, Paus T, Pausova Z. Obesity and the cerebral cortex: Underlying neurobiology in mice and humans. Brain Behav Immun 2024; 119:637-647. [PMID: 38663773 DOI: 10.1016/j.bbi.2024.04.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/01/2024] Open
Abstract
Obesity is a major modifiable risk factor for Alzheimer's disease (AD), characterized by progressive atrophy of the cerebral cortex. The neurobiology of obesity contributions to AD is poorly understood. Here we show with in vivo MRI that diet-induced obesity decreases cortical volume in mice, and that higher body adiposity associates with lower cortical volume in humans. Single-nuclei transcriptomics of the mouse cortex reveals that dietary obesity promotes an array of neuron-adverse transcriptional dysregulations, which are mediated by an interplay of excitatory neurons and glial cells, and which involve microglial activation and lowered neuronal capacity for neuritogenesis and maintenance of membrane potential. The transcriptional dysregulations of microglia, more than of other cell types, are like those in AD, as assessed with single-nuclei cortical transcriptomics in a mouse model of AD and two sets of human donors with the disease. Serial two-photon tomography of microglia demonstrates microgliosis throughout the mouse cortex. The spatial pattern of adiposity-cortical volume associations in human cohorts interrogated together with in silico bulk and single-nucleus transcriptomic data from the human cortex implicated microglia (along with other glial cells and subtypes of excitatory neurons), and it correlated positively with the spatial profile of cortical atrophy in patients with mild cognitive impairment and AD. Thus, multi-cell neuron-adverse dysregulations likely contribute to the loss of cortical tissue in obesity. The dysregulations of microglia may be pivotal to the obesity-related risk of AD.
Collapse
Affiliation(s)
- Yash Patel
- The Hospital for Sick Children, Translational Medicine Program, Toronto, ON, Canada; Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Anita Woo
- The Hospital for Sick Children, Translational Medicine Program, Toronto, ON, Canada; Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Sammy Shi
- The Hospital for Sick Children, Translational Medicine Program, Toronto, ON, Canada; Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Ramy Ayoub
- The Hospital for Sick Children, Translational Medicine Program, Toronto, ON, Canada
| | - Jean Shin
- The Hospital for Sick Children, Translational Medicine Program, Toronto, ON, Canada; Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Amy Botta
- The Hospital for Sick Children, Translational Medicine Program, Toronto, ON, Canada
| | - Troy Ketela
- Princess Margaret Genomics Centre, Toronto, ON, Canada
| | - Hoon-Ki Sung
- The Hospital for Sick Children, Translational Medicine Program, Toronto, ON, Canada
| | - Jason Lerch
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, Great Britton
| | - Brian Nieman
- The Hospital for Sick Children, Translational Medicine Program, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Tomas Paus
- Department of Psychiatry and Addictology and Department of Neuroscience, Faculty of Medicine and Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, QC, Canada
| | - Zdenka Pausova
- The Hospital for Sick Children, Translational Medicine Program, Toronto, ON, Canada; Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada; Department of Pediatrics and Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, QC, Canada.
| |
Collapse
|
22
|
Islam MA, Sehar U, Sultana OF, Mukherjee U, Brownell M, Kshirsagar S, Reddy PH. SuperAgers and centenarians, dynamics of healthy ageing with cognitive resilience. Mech Ageing Dev 2024; 219:111936. [PMID: 38657874 DOI: 10.1016/j.mad.2024.111936] [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: 03/08/2024] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024]
Abstract
Graceful healthy ageing and extended longevity is the most desired goal for human race. The process of ageing is inevitable and has a profound impact on the gradual deterioration of our physiology and health since it triggers the onset of many chronic conditions like dementia, osteoporosis, diabetes, arthritis, cancer, and cardiovascular disease. However, some people who lived/live more than 100 years called 'Centenarians" and how do they achieve their extended lifespans are not completely understood. Studying these unknown factors of longevity is important not only to establish a longer human lifespan but also to manage and treat people with shortened lifespans suffering from age-related morbidities. Furthermore, older adults who maintain strong cognitive function are referred to as "SuperAgers" and may be resistant to risk factors linked to cognitive decline. Investigating the mechanisms underlying their cognitive resilience may contribute to the development of therapeutic strategies that support the preservation of cognitive function as people age. The key to a long, physically, and cognitively healthy life has been a mystery to scientists for ages. Developments in the medical sciences helps us to a better understanding of human physiological function and greater access to medical care has led us to an increase in life expectancy. Moreover, inheriting favorable genetic traits and adopting a healthy lifestyle play pivotal roles in promoting longer and healthier lives. Engaging in regular physical activity, maintaining a balanced diet, and avoiding harmful habits such as smoking contribute to overall well-being. The synergy between positive lifestyle choices, access to education, socio-economic factors, environmental determinants and genetic supremacy enhances the potential for a longer and healthier life. Our article aims to examine the factors associated with healthy ageing, particularly focusing on cognitive health in centenarians. We will also be discussing different aspects of ageing including genomic instability, metabolic burden, oxidative stress and inflammation, mitochondrial dysfunction, cellular senescence, immunosenescence, and sarcopenia.
Collapse
Affiliation(s)
- Md Ariful Islam
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Omme Fatema Sultana
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Upasana Mukherjee
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Malcolm Brownell
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX 79409, USA.
| |
Collapse
|
23
|
Pandit H, Jones NS, Rebeck GW. Obesity affects brain cortex gene expression in an APOE genotype and sex dependent manner. Int J Obes (Lond) 2024; 48:841-848. [PMID: 38454009 PMCID: PMC11379128 DOI: 10.1038/s41366-024-01481-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 01/07/2024] [Accepted: 01/19/2024] [Indexed: 03/09/2024]
Abstract
OBJECTIVE Obesity is the top modifiable risk factor for Alzheimer's disease. We hypothesized that high fat diet (HFD)-induced obesity alters brain transcriptomics in APOE-genotype and sex dependent manners. Here, we investigated interactions between HFD, APOE, and sex, using a knock-in mouse model of the human APOE3 and APOE4 alleles. METHODS Six-month-old APOE3-TR and APOE4-TR mice were treated with either HFD or control chow. After 4 months, total RNA was extracted from the cerebral cortices and analyzed by poly-A enriched RNA sequencing on the Illumina platform. RESULTS Female mice demonstrated profound HFD-induced transcriptomic changes while there was little to no effect in males. In females, APOE3 brains demonstrated about five times more HFD-induced transcriptomic changes (399 up-regulated and 107 down-regulated genes) compared to APOE4 brains (30 up-regulated and 60 down-regulated). Unsupervised clustering analysis revealed two gene sets that responded to HFD in APOE3 mice but not in APOE4 mice. Pathway analysis demonstrated that HFD in APOE3 mice affected cortical pathways related to feeding behavior, blood circulation, circadian rhythms, extracellular matrix, and cell adhesion. CONCLUSIONS Female mice and APOE3 mice have the strongest cortical transcriptomic responses to HFD related to feeding behavior and extracellular matrix remodeling. The relative lack of response of the APOE4 brain to stress associated with obesity may leave it more susceptible to additional stresses that occur with aging and in AD.
Collapse
Affiliation(s)
- Harshul Pandit
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Road N.W., Washington, DC, 20007, USA
| | - Nahdia S Jones
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Road N.W., Washington, DC, 20007, USA
| | - G William Rebeck
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Road N.W., Washington, DC, 20007, USA.
| |
Collapse
|
24
|
Poxleitner M, Hoffmann SHL, Berezhnoy G, Ionescu TM, Gonzalez-Menendez I, Maier FC, Seyfried D, Ehrlichmann W, Quintanilla-Martinez L, Schmid AM, Reischl G, Trautwein C, Maurer A, Pichler BJ, Herfert K, Beziere N. Western diet increases brain metabolism and adaptive immune responses in a mouse model of amyloidosis. J Neuroinflammation 2024; 21:129. [PMID: 38745337 PMCID: PMC11092112 DOI: 10.1186/s12974-024-03080-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/29/2024] [Indexed: 05/16/2024] Open
Abstract
Diet-induced increase in body weight is a growing health concern worldwide. Often accompanied by a low-grade metabolic inflammation that changes systemic functions, diet-induced alterations may contribute to neurodegenerative disorder progression as well. This study aims to non-invasively investigate diet-induced metabolic and inflammatory effects in the brain of an APPPS1 mouse model of Alzheimer's disease. [18F]FDG, [18F]FTHA, and [18F]GE-180 were used for in vivo PET imaging in wild-type and APPPS1 mice. Ex vivo flow cytometry and histology in brains complemented the in vivo findings. 1H- magnetic resonance spectroscopy in the liver, plasma metabolomics and flow cytometry of the white adipose tissue were used to confirm metaflammatory condition in the periphery. We found disrupted glucose and fatty acid metabolism after Western diet consumption, with only small regional changes in glial-dependent neuroinflammation in the brains of APPPS1 mice. Further ex vivo investigations revealed cytotoxic T cell involvement in the brains of Western diet-fed mice and a disrupted plasma metabolome. 1H-magentic resonance spectroscopy and immunological results revealed diet-dependent inflammatory-like misbalance in livers and fatty tissue. Our multimodal imaging study highlights the role of the brain-liver-fat axis and the adaptive immune system in the disruption of brain homeostasis in amyloid models of Alzheimer's disease.
Collapse
Affiliation(s)
- Marilena Poxleitner
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Sabrina H L Hoffmann
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Georgy Berezhnoy
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Tudor M Ionescu
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Irene Gonzalez-Menendez
- Department of Pathology and Neuropathology, University Hospital Tübingen, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Florian C Maier
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Dominik Seyfried
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Walter Ehrlichmann
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Leticia Quintanilla-Martinez
- Department of Pathology and Neuropathology, University Hospital Tübingen, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Andreas M Schmid
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Gerald Reischl
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Christoph Trautwein
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Andreas Maurer
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Kristina Herfert
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany.
| | - Nicolas Beziere
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany.
- Cluster of Excellence CMFI (EXC 2124) "Controlling Microbes to Fight Infections", Eberhard Karls University, Tübingen, Germany.
| |
Collapse
|
25
|
Evans AK, Saw NL, Woods CE, Vidano LM, Blumenfeld SE, Lam RK, Chu EK, Reading C, Shamloo M. Impact of high-fat diet on cognitive behavior and central and systemic inflammation with aging and sex differences in mice. Brain Behav Immun 2024; 118:334-354. [PMID: 38408498 PMCID: PMC11019935 DOI: 10.1016/j.bbi.2024.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 02/28/2024] Open
Abstract
Aging and age-related diseases are associated with cellular stress, metabolic imbalance, oxidative stress, and neuroinflammation, accompanied by cognitive impairment. Lifestyle factors such as diet, sleep fragmentation, and stress can potentiate damaging cellular cascades and lead to an acceleration of brain aging and cognitive impairment. High-fat diet (HFD) has been associated with obesity, metabolic disorders like diabetes, and cardiovascular disease. HFD also induces neuroinflammation, impairs learning and memory, and may increase anxiety-like behavior. Effects of a HFD may also vary between sexes. The interaction between Age- and Sex- and Diet-related changes in neuroinflammation and cognitive function is an important and poorly understood area of research. This study was designed to examine the effects of HFD on neuroinflammation, behavior, and neurodegeneration in mice in the context of aging or sex differences. In a series of studies, young (2-3 months) or old (12-13 months) C57BL/6J male mice or young male and female C57Bl/6J mice were fed either a standard diet (SD) or a HFD for 5-6 months. Behavior was assessed in Activity Chamber, Y-maze, Novel Place Recognition, Novel Object Recognition, Elevated Plus Maze, Open Field, Morris Water Maze, and Fear Conditioning. Post-mortem analyses assessed a panel of inflammatory markers in the plasma and hippocampus. Additionally, proteomic analysis of the hypothalamus, neurodegeneration, neuroinflammation in the locus coeruleus, and neuroinflammation in the hippocampus were assessed in a subset of young and aged male mice. We show that HFD increased body weight and decreased locomotor activity across groups compared to control mice fed a SD. HFD altered anxiety-related exploratory behavior. HFD impaired spatial learning and recall in young male mice and impaired recall in cued fear conditioning in young and aged male mice, with no effects on spatial learning or fear conditioning in young female mice. Effects of Age and Sex were observed on neuroinflammatory cytokines, with only limited effects of HFD. HFD had a more significant impact on systemic inflammation in plasma across age and sex. Aged male mice had induction of microglial immunoreactivity in both the locus coeruleus (LC) and hippocampus an effect that HFD exacerbated in the hippocampal CA1 region. Proteomic analysis of the hypothalamus revealed changes in pathways related to metabolism and neurodegeneration with both aging and HFD in male mice. Our findings suggest that HFD induces widespread systemic inflammation and limited neuroinflammation. In addition, HFD alters exploratory behavior in male and female mice, and impairs learning and memory in male mice. These results provide valuable insight into the impact of diet on cognition and aging pathophysiology.
Collapse
Affiliation(s)
- Andrew K Evans
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | - Nay L Saw
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | - Claire E Woods
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | - Laura M Vidano
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | - Sarah E Blumenfeld
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | - Rachel K Lam
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | - Emily K Chu
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | | | - Mehrdad Shamloo
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304.
| |
Collapse
|
26
|
Balasubramanian P, Kiss T, Gulej R, Nyul Toth A, Tarantini S, Yabluchanskiy A, Ungvari Z, Csiszar A. Accelerated Aging Induced by an Unhealthy High-Fat Diet: Initial Evidence for the Role of Nrf2 Deficiency and Impaired Stress Resilience in Cellular Senescence. Nutrients 2024; 16:952. [PMID: 38612986 PMCID: PMC11013792 DOI: 10.3390/nu16070952] [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: 11/22/2023] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
High-fat diets (HFDs) have pervaded modern dietary habits, characterized by their excessive saturated fat content and low nutritional value. Epidemiological studies have compellingly linked HFD consumption to obesity and the development of type 2 diabetes mellitus. Moreover, the synergistic interplay of HFD, obesity, and diabetes expedites the aging process and prematurely fosters age-related diseases. However, the underlying mechanisms driving these associations remain enigmatic. One of the most conspicuous hallmarks of aging is the accumulation of highly inflammatory senescent cells, with mounting evidence implicating increased cellular senescence in the pathogenesis of age-related diseases. Our hypothesis posits that HFD consumption amplifies senescence burden across multiple organs. To scrutinize this hypothesis, we subjected mice to a 6-month HFD regimen, assessing senescence biomarker expression in the liver, white adipose tissue, and the brain. Aging is intrinsically linked to impaired cellular stress resilience, driven by dysfunction in Nrf2-mediated cytoprotective pathways that safeguard cells against oxidative stress-induced senescence. To ascertain whether Nrf2-mediated pathways shield against senescence induction in response to HFD consumption, we explored senescence burden in a novel model of aging: Nrf2-deficient (Nrf2+/-) mice, emulating the aging phenotype. Our initial findings unveiled significant Nrf2 dysfunction in Nrf2+/- mice, mirroring aging-related alterations. HFD led to substantial obesity, hyperglycemia, and impaired insulin sensitivity in both Nrf2+/- and Nrf2+/+ mice. In control mice, HFD primarily heightened senescence burden in white adipose tissue, evidenced by increased Cdkn2a senescence biomarker expression. In Nrf2+/- mice, HFD elicited a significant surge in senescence burden across the liver, white adipose tissue, and the brain. We postulate that HFD-induced augmentation of senescence burden may be a pivotal contributor to accelerated organismal aging and the premature onset of age-related diseases.
Collapse
Affiliation(s)
- Priya Balasubramanian
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Tamas Kiss
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
- International Training Program in Geroscience, First Department of Pediatrics, Semmelweis University, 1089 Budapest, Hungary
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Adam Nyul Toth
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
| |
Collapse
|
27
|
Opęchowska A, Karpiuk K, Zahorodnii A, Harasim-Symbor E, Chabowski A, Konstantynowicz-Nowicka K. Anti-inflammatory effects of cannabidiol in early stages of neuroinflammation induced by high-fat diet in cerebral cortex of rats. Toxicol Appl Pharmacol 2024; 484:116856. [PMID: 38336253 DOI: 10.1016/j.taap.2024.116856] [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: 10/22/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
High-fat diet (HFD) contributes to neuroinflammation forming, hence it is crucial to find safe and effective substances that are able to counteract its progress. The anti-inflammatory properties of phytocannabinoids acquired from the Cannabis plant have been widely acknowledged. We evaluated the effects of cannabidiol (CBD) treatment on induced by applying HFD early stages of neuroinflammation in Wistar rat cerebral cortex. In our 7-week experiment, CBD was injected intraperitoneally over the last 14days at a dose of 10 mg/kg of body weight once a day. The level of arachidonic acid, a precursor to pro-inflammatory eicosanoids, decreased in all analysed lipid classes after CBD administration to the HFD group. Moreover, the extent of diminishing the activity of the omega-6 (n-6) fatty acid pathway by CBD was the greatest in diacylglycerols and phospholipids. Surprisingly, CBD was also capable of downregulating the activity of the omega-3 (n-3) pathway. The expression of enzymes involved in the synthesis of the eicosanoids was significantly increased in the HFD group and subsequently lowered by CBD. Significant changes in various cytokines levels were also discovered. Our results strongly suggest the ability of CBD to reduce the formation of lipid inflammation precursors in rat cerebral cortex, as a primary event in the development of neurodegenerative diseases. This can raise hopes for the future use of this cannabinoid for therapeutic purposes since it is a substance lacking lasting and severe side effects.
Collapse
Affiliation(s)
- Aleksandra Opęchowska
- Department of Physiology, Medical University of Bialystok, Mickiewicz Str. 2C, Bialystok 15-222, Poland.
| | - Kacper Karpiuk
- Department of Physiology, Medical University of Bialystok, Mickiewicz Str. 2C, Bialystok 15-222, Poland.
| | - Andrii Zahorodnii
- Department of Physiology, Medical University of Bialystok, Mickiewicz Str. 2C, Bialystok 15-222, Poland.
| | - Ewa Harasim-Symbor
- Department of Physiology, Medical University of Bialystok, Mickiewicz Str. 2C, Bialystok 15-222, Poland.
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Mickiewicz Str. 2C, Bialystok 15-222, Poland.
| | | |
Collapse
|
28
|
Sanchez C, Colson C, Gautier N, Noser P, Salvi J, Villet M, Fleuriot L, Peltier C, Schlich P, Brau F, Sharif A, Altintas A, Amri EZ, Nahon JL, Blondeau N, Benani A, Barrès R, Rovère C. Dietary fatty acid composition drives neuroinflammation and impaired behavior in obesity. Brain Behav Immun 2024; 117:330-346. [PMID: 38309640 DOI: 10.1016/j.bbi.2024.01.216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/17/2024] [Accepted: 01/20/2024] [Indexed: 02/05/2024] Open
Abstract
Nutrient composition in obesogenic diets may influence the severity of disorders associated with obesity such as insulin-resistance and chronic inflammation. Here we hypothesized that obesogenic diets rich in fat and varying in fatty acid composition, particularly in omega 6 (ω6) to omega 3 (ω3) ratio, have various effects on energy metabolism, neuroinflammation and behavior. Mice were fed either a control diet or a high fat diet (HFD) containing either low (LO), medium (ME) or high (HI) ω6/ω3 ratio. Mice from the HFD-LO group consumed less calories and exhibited less body weight gain compared to other HFD groups. Both HFD-ME and HFD-HI impaired glucose metabolism while HFD-LO partly prevented insulin intolerance and was associated with normal leptin levels despite higher subcutaneous and perigonadal adiposity. Only HFD-HI increased anxiety and impaired spatial memory, together with increased inflammation in the hypothalamus and hippocampus. Our results show that impaired glucose metabolism and neuroinflammation are uncoupled, and support that diets with a high ω6/ω3 ratio are associated with neuroinflammation and the behavioral deterioration coupled with the consumption of diets rich in fat.
Collapse
Affiliation(s)
- Clara Sanchez
- Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, France
| | - Cécilia Colson
- Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, France; Université Côte d'Azur, Institut de Biologie de Valrose, CNRS, INSERM, France
| | - Nadine Gautier
- Université Côte d'Azur, Institut de Biologie de Valrose, CNRS, INSERM, France
| | - Pascal Noser
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Denmark
| | - Juliette Salvi
- Université Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAe, France
| | - Maxime Villet
- Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, France
| | - Lucile Fleuriot
- Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, France
| | - Caroline Peltier
- Université Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAe, France
| | - Pascal Schlich
- Université Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAe, France
| | - Frédéric Brau
- Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, France
| | - Ariane Sharif
- Université de Lille, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neurosciences & Cognition, UMR-S 1172, Lille France
| | - Ali Altintas
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Denmark
| | - Ez-Zoubir Amri
- Université Côte d'Azur, Institut de Biologie de Valrose, CNRS, INSERM, France
| | - Jean-Louis Nahon
- Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, France
| | - Nicolas Blondeau
- Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, France
| | - Alexandre Benani
- Université Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAe, France
| | - Romain Barrès
- Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, France; Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Denmark
| | - Carole Rovère
- Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, France.
| |
Collapse
|
29
|
Liu J, Zaidi A, Pike CJ. Microglia/macrophage-specific deletion of TLR-4 protects against neural effects of diet-induced obesity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.13.580189. [PMID: 38405877 PMCID: PMC10888944 DOI: 10.1101/2024.02.13.580189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Obesity is associated with numerous adverse neural effects, including reduced neurogenesis, cognitive impairment, and increased risks for developing Alzheimer's disease (AD) and vascular dementia. Obesity is also characterized by chronic, low-grade inflammation that is implicated in mediating negative consequences body-wide. Toll-like receptor 4 (TLR4) signaling from peripheral macrophages is implicated as an essential regulator of the systemic inflammatory effects of obesity. In the brain, obesity drives chronic neuroinflammation that involves microglial activation, however the contributions of microglia-derived TLR4 signaling to the consequences of obesity are poorly understood. To investigate this issue, we first generated mice that carry an inducible, microglia/macrophage-specific deletion of TLR4 that yields long-term TLR4 knockout only in brain indicating microglial specificity. Next, we analyzed the effects of microglial TLR4 deletion on systemic and neural effects of a 16-week of exposure to control versus obesogenic high-fat diets. In male mice, TLR4 deletion generally yielded limited effects on diet-induced systemic metabolic dysfunction but significantly reduced neuroinflammation and impairments in neurogenesis and cognitive performance. In female mice maintained on obesogenic diet, TLR4 deletion partially protected against weight gain, adiposity, and metabolic impairments. Compared to males, females showed milder diet-induced neural consequences, against which TLR4 deletion was protective. Collectively, these findings demonstrate a central role of microglial TLR4 signaling in mediating the neural effects of obesogenic diet and highlight sexual dimorphic responses to both diet and TLR4.
Collapse
|
30
|
Rossi C, Distaso M, Raggi F, Kusmic C, Faita F, Solini A. Lacking P2X7-receptors protects substantia nigra dopaminergic neurons and hippocampal-related cognitive performance from the deleterious effects of high-fat diet exposure in adult male mice. Front Nutr 2024; 11:1289750. [PMID: 38344021 PMCID: PMC10854005 DOI: 10.3389/fnut.2024.1289750] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/11/2024] [Indexed: 01/05/2025] Open
Abstract
BACKGROUND Dietary fat consumption, involved in the pathogenesis of insulin resistance and impaired glucose metabolism, is linked with decline in cognitive functions, dementia, and development of Parkinson's disease and Alzheimer's disease. Mature IL-1β, requiring the activation of the P2X7 receptor (P2X7R)-inflammasome complex, is an important mediator of neuroinflammation. The aim of the study was to test whether P2X7R activation might interfere with systemic and cerebral metabolic homeostasis. METHODS We treated WT and P2X7R KO mice with a high-fat diet (HFD) for 16 weeks, evaluating the effects on the Substantia Nigra and Hippocampus, target areas of damage in several forms of cognitive impairment. RESULTS HFD-treated WT and P2X7R KO mice showed a different brain mRNA profile of Insulin and Igf-1, with these genes and relative receptors, more expressed in KO mice. Unlike P2X7R KO mice, WT mice treated with HFD displayed a diameter reduction in dopaminergic neurons in the Substantia Nigra, accompanied by an increased IBA1 expression in this area; they also showed poor performances during Y-Maze and Morris Water Maze, tasks involving Hippocampus activity. Conversely, Parkin, whose reduction might promote neuronal cell death, was increased in the brain of P2X7R KO animals. CONCLUSION We report for the first time that HFD induces damage in dopaminergic neurons of the Substantia Nigra and a Hippocampus-related worse cognitive performance, both attenuated in the absence of P2X7R. The involved mechanisms might differ in the two brain areas, with a predominant role of inflammation in the Substantia Nigra and a metabolic derangement in the Hippocampus.
Collapse
Affiliation(s)
- Chiara Rossi
- Department of Surgical, Medical, Molecular and Critical Area Pathology University of Pisa, Pisa, Italy
| | - Mariarosaria Distaso
- Department of Surgical, Medical, Molecular and Critical Area Pathology University of Pisa, Pisa, Italy
| | - Francesco Raggi
- Department of Surgical, Medical, Molecular and Critical Area Pathology University of Pisa, Pisa, Italy
| | | | | | - Anna Solini
- Department of Surgical, Medical, Molecular and Critical Area Pathology University of Pisa, Pisa, Italy
| |
Collapse
|
31
|
Baer SB, Dorn AD, Osborne DM. Sex differences in response to obesity and caloric restriction on cognition and hippocampal measures of autophagic-lysosomal transcripts and signaling pathways. BMC Neurosci 2024; 25:1. [PMID: 38166559 PMCID: PMC10759648 DOI: 10.1186/s12868-023-00840-1] [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: 04/20/2023] [Accepted: 12/18/2023] [Indexed: 01/04/2024] Open
Abstract
BACKGROUND Obesity rates in the U.S. continue to increase, with nearly 50% of the population being either obese or morbidly obese. Obesity, along with female sex, are leading risk factors for sporadic Alzheimer's Disease (AD) necessitating the need to better understand how these variables impact cellular function independent of age or genetic mutations. Animal and clinical studies both indicate that autophagy-lysosomal pathway (ALP) dysfunction is among the earliest known cellular systems to become perturbed in AD, preceding cognitive decline, yet little is known about how obesity and sex affects these cellular functions in the hippocampus, a brain region uniquely susceptible to the negative effects of obesity. We hypothesized that obesity would negatively affect key markers of ALP in the hippocampus, effects would vary based on sex, and that caloric restriction would counteract obesity effects. METHODS Female and male mice were placed on an obesogenic diet for 10 months, at which point half were switched to caloric restriction for three months, followed by cognitive testing in the Morris watermaze. Hippocampus was analyzed by western blot and qPCR. RESULTS Cognitive function in female mice responded differently to caloric restriction based on whether they were on a normal or obesogenic diet; male cognition was only mildly affected by caloric restriction and not obesity. Significant male-specific changes occurred in cellular markers of autophagy, including obesity increasing pAkt, Slc38a9, and Atg12, while caloric restriction reduced pRPS6 and increased Atg7. In contrast females experienced changes due to diet/caloric restriction predominately in lysosomal markers including increased TFE3, FLCN, FNIP2, and pAMPK. CONCLUSIONS Results support that hippocampal ALP is a target of obesity and that sex shapes molecular responses, while providing insight into how dietary manipulations affect learning and memory based on sex.
Collapse
Affiliation(s)
- Sadie B Baer
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR, USA
| | - Adrianah D Dorn
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR, USA
| | | |
Collapse
|
32
|
Viguier C, Bullich S, Botella M, Fasseu L, Alfonso A, Rekik K, Gauzin S, Guiard BP, Davezac N. Impact of physical activity on brain oxidative metabolism and intrinsic capacities in young swiss mice fed a high fat diet. Neuropharmacology 2023; 241:109730. [PMID: 37758019 DOI: 10.1016/j.neuropharm.2023.109730] [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: 03/31/2023] [Revised: 09/22/2023] [Accepted: 09/23/2023] [Indexed: 10/03/2023]
Abstract
Type 2 diabetes and obesity characterized by hallmarks of insulin resistance along with an imbalance in brain oxidative metabolism would impair intrinsic capacities (ICs), a new concept for assessing mental and physical functioning. Here, we explored the impact of physical activity on antioxidant responses and oxidative metabolism in discrete brain areas of HFD or standard diet (STD) fed mice but also its consequences on specific domains of ICs. 6-week-old Swiss male mice were exposed to a STD or a HFD for 16 weeks and half of the mice in each group had access to an activity wheel and the other half did not. As expected HFD mice displayed peripheral insulin resistance but also a persistent inhibition of aconitase activity in cortices revealing an increase in mitochondrial reactive oxygen species (ROS) production. Animals with access to the running wheel displayed an improvement of insulin sensitivity regardless of the diet factor whereas ROS production remained impaired. Moreover, although the access of the running wheel did not influence mitochondrial biomass, in the oxidative metabolism area, it produced a slight decrease in brain SOD1 and catalase expression notably in HFD fed mice. At the behavioural level, physical exercise produced anxiolytic/antidepressant-like responses and improved motor coordination in both STD and HFD fed mice. However, this non-pharmacological intervention failed to enhance cognitive performance. These findings paint a contrasting landscape about physical exercise as a non-pharmacological intervention for positively orienting the aging trajectory.
Collapse
Affiliation(s)
- Clémence Viguier
- Remember Team, Research Center on Animal Cognition (CRCA), Center of Integrative Biology (CBI), CNRS - University of Toulouse, CNRS, UPS, 31 067, Toulouse, France
| | - Sébastien Bullich
- Remember Team, Research Center on Animal Cognition (CRCA), Center of Integrative Biology (CBI), CNRS - University of Toulouse, CNRS, UPS, 31 067, Toulouse, France
| | - Marlene Botella
- Minding Team, Research Center on Animal Cognition (CRCA), Center of Integrative Biology (CBI), CNRS - University of Toulouse, CNRS, UPS, 31 067, Toulouse, France; INSPIRE Consortium, France
| | - Laure Fasseu
- Minding Team, Research Center on Animal Cognition (CRCA), Center of Integrative Biology (CBI), CNRS - University of Toulouse, CNRS, UPS, 31 067, Toulouse, France; INSPIRE Consortium, France
| | - Amélie Alfonso
- Remember Team, Research Center on Animal Cognition (CRCA), Center of Integrative Biology (CBI), CNRS - University of Toulouse, CNRS, UPS, 31 067, Toulouse, France; INSPIRE Consortium, France
| | - Khaoula Rekik
- Remember Team, Research Center on Animal Cognition (CRCA), Center of Integrative Biology (CBI), CNRS - University of Toulouse, CNRS, UPS, 31 067, Toulouse, France
| | - Sébastien Gauzin
- Remember Team, Research Center on Animal Cognition (CRCA), Center of Integrative Biology (CBI), CNRS - University of Toulouse, CNRS, UPS, 31 067, Toulouse, France; INSPIRE Consortium, France
| | - Bruno P Guiard
- Remember Team, Research Center on Animal Cognition (CRCA), Center of Integrative Biology (CBI), CNRS - University of Toulouse, CNRS, UPS, 31 067, Toulouse, France; INSPIRE Consortium, France.
| | - Noélie Davezac
- Minding Team, Research Center on Animal Cognition (CRCA), Center of Integrative Biology (CBI), CNRS - University of Toulouse, CNRS, UPS, 31 067, Toulouse, France; INSPIRE Consortium, France.
| |
Collapse
|
33
|
Hung KC, Liu CC, Wu JY, Ho CN, Lin MC, Hsing CH, Chen IW. Association between the neutrophil-to-lymphocyte ratio and cognitive impairment: a meta-analysis of observational studies. Front Endocrinol (Lausanne) 2023; 14:1265637. [PMID: 38089627 PMCID: PMC10715314 DOI: 10.3389/fendo.2023.1265637] [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: 07/23/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
Background Systemic inflammation is one of the underlying mechanisms of cognitive impairment. The neutrophil-to-lymphocyte ratio (NLR) has emerged as a systemic inflammation indicator. This meta-analysis aimed to evaluate the association between high NLR and cognitive impairment (CI) risk. Method A comprehensive systematic search was conducted to identify eligible studies published until May 30, 2023. The reference group comprised patients with the lowest NLR level, whereas the exposure group comprised those with the highest NLR level. The main outcome was to examine the relationship between NLR and CI risk. The secondary outcome included the association between patient characteristics or comorbidities and CI risk. Results This meta-analysis included 11 studies published between 2018 and 2023, involving 10,357 patients. Patients with CI had a higher NLR than those without (mean difference=0.35, 95% confidence interval [CI]: 0.26-0.44, p < 00001, I2 = 86%). Consistently, pooled results revealed an association between high NLR and CI risk (odds ratio [OR]=2.53, 95% CI:1.67-3.82, p<0.0001, I2 = 84%). Furthermore, aging (mean difference =4.31 years, 95% CI:2.83-5.8, p < 0.00001, I2 = 92%), diabetes (OR=1.59, 95% CI:1.35-1.88, p < 0.00001, I2 = 66%), and hypertension (OR=1.36, 95% CI:1.19-1.57, p < 0.00001, I2 = 0%) were significant risk factors for CI. However, no significant associations were observed between CI and male gender (OR = 0.84, 95% CI:0.64-1.11, p = 0.22, I2 = 81%), body mass index (mean = -0.32 kg/m2, 95% CI: -0.82, 0.18, p = 0.2, I2 = 82%), alcohol consumption (OR = 1.11, 95% CI:0.95-1.3, p = 1.35, I2 = 0%), and smoking (OR = 0.99, 95% CI:0.87-1.13, p = 0.86, I2 = 0%). Meta-regression found that diabetes and hypertension, but not age, significantly moderated the association between NLR and CI. Conclusion This meta-analysis showed a significant association between high NLR and increased CI risk. Moreover, meta-regression identified diabetes and hypertension, but not age, as significant moderating factors in the relationship between NLR and CI. To validate and strengthen these findings, further large-scale studies are required. Systematic Review Registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42023430384, identifier CRD42023430384.
Collapse
Affiliation(s)
- Kuo-Chuan Hung
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Anesthesiology, Chi Mei Medical Center, Tainan, Taiwan
| | - Chien-Cheng Liu
- Department of Anesthesiology, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
- Department of Nursing, College of Medicine, I-Shou University, Kaohsiung, Taiwan
- School of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Jheng-Yan Wu
- Department of Nutrition, Chi Mei Medical Center, Tainan, Taiwan
| | - Chun-Ning Ho
- Department of Anesthesiology, Chi Mei Medical Center, Tainan, Taiwan
| | - Ming-Chung Lin
- Department of Anesthesiology, Chi Mei Medical Center, Tainan, Taiwan
| | - Chung-Hsi Hsing
- Department of Anesthesiology, Chi Mei Medical Center, Tainan, Taiwan
- Department of Medical Research, Chi-Mei Medical Center, Tainan, Taiwan
| | - I-Wen Chen
- Department of Anesthesiology, Chi Mei Medical Center, Liouying, Tainan, Taiwan
| |
Collapse
|
34
|
Shetty S, Duesman SJ, Patel S, Huyhn P, Shroff S, Das A, Chowhan D, Sebra R, Beaumont K, McAlpine CS, Rajbhandari P, Rajbhandari AK. Sexually dimorphic role of diet and stress on behavior, energy metabolism, and the ventromedial hypothalamus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.567534. [PMID: 38014350 PMCID: PMC10680837 DOI: 10.1101/2023.11.17.567534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Scientific evidence underscores the influence of biological sex on the interplay between stress and metabolic dysfunctions. However, there is limited understanding of how diet and stress jointly contribute to metabolic dysregulation in both males and females. To address this gap, our study aimed to investigate the combined effects of a high-fat diet (HFD) and repeated footshock stress on fear-related behaviors and metabolic outcomes in male and female mice. Using a robust rodent model that recapitulates key aspects of post-traumatic stress disorder (PTSD), we subjected mice to footshock stressor followed by weekly reminder footshock stressor or no stressor for 14 weeks while on either an HFD or chow diet. Our findings revealed that HFD impaired fear memory extinction in male mice that received initial stressor but not in female mice. Blood glucose levels were influenced by both diet and sex, with HFD-fed female mice displaying elevated levels that returned to baseline in the absence of stress, a pattern not observed in male mice. Male mice on HFD exhibited higher energy expenditure, while HFD-fed female mice showed a decreased respiratory exchange ratio (RER). Sex-specific alterations in pro-inflammatory markers and abundance of hematopoietic stem cells were observed in chronically stressed mice on an HFD in different peripheral tissues, indicating the manifestation of distinct comorbid disorders. Single-nuclei RNA sequencing of the ventromedial hypothalamus from stressed mice on an HFD provided insights into sex-specific glial cell activation and cell-type-specific transcriptomic changes. In conclusion, our study offers a comprehensive understanding of the intricate interactions between stress, diet, sex, and various physiological and behavioral outcomes, shedding light on a potential brain region coordinating these interactions.
Collapse
Affiliation(s)
- Sanutha Shetty
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Samuel J. Duesman
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Sanil Patel
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Pacific Huyhn
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Sanjana Shroff
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anika Das
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
- Center for Excellence in Youth Education, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Disha Chowhan
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Sebra
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristin Beaumont
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cameron S. McAlpine
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Prashant Rajbhandari
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, NY, New York 10029
- Disease Mechanism and Therapeutics Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Senior authors
| | - Abha K. Rajbhandari
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
- Senior authors
| |
Collapse
|
35
|
de las Fuentes L, Schwander KL, Brown MR, Bentley AR, Winkler TW, Sung YJ, Munroe PB, Miller CL, Aschard H, Aslibekyan S, Bartz TM, Bielak LF, Chai JF, Cheng CY, Dorajoo R, Feitosa MF, Guo X, Hartwig FP, Horimoto A, Kolčić I, Lim E, Liu Y, Manning AK, Marten J, Musani SK, Noordam R, Padmanabhan S, Rankinen T, Richard MA, Ridker PM, Smith AV, Vojinovic D, Zonderman AB, Alver M, Boissel M, Christensen K, Freedman BI, Gao C, Giulianini F, Harris SE, He M, Hsu FC, Kühnel B, Laguzzi F, Li X, Lyytikäinen LP, Nolte IM, Poveda A, Rauramaa R, Riaz M, Robino A, Sofer T, Takeuchi F, Tayo BO, van der Most PJ, Verweij N, Ware EB, Weiss S, Wen W, Yanek LR, Zhan Y, Amin N, Arking DE, Ballantyne C, Boerwinkle E, Brody JA, Broeckel U, Campbell A, Canouil M, Chai X, Chen YDI, Chen X, Chitrala KN, Concas MP, de Faire U, de Mutsert R, de Silva HJ, de Vries PS, Do A, Faul JD, Fisher V, Floyd JS, Forrester T, Friedlander Y, Girotto G, Gu CC, Hallmans G, Heikkinen S, Heng CK, Homuth G, Hunt S, Ikram MA, Jacobs DR, Kavousi M, Khor CC, Kilpeläinen TO, Koh WP, Komulainen P, Langefeld CD, Liang J, et alde las Fuentes L, Schwander KL, Brown MR, Bentley AR, Winkler TW, Sung YJ, Munroe PB, Miller CL, Aschard H, Aslibekyan S, Bartz TM, Bielak LF, Chai JF, Cheng CY, Dorajoo R, Feitosa MF, Guo X, Hartwig FP, Horimoto A, Kolčić I, Lim E, Liu Y, Manning AK, Marten J, Musani SK, Noordam R, Padmanabhan S, Rankinen T, Richard MA, Ridker PM, Smith AV, Vojinovic D, Zonderman AB, Alver M, Boissel M, Christensen K, Freedman BI, Gao C, Giulianini F, Harris SE, He M, Hsu FC, Kühnel B, Laguzzi F, Li X, Lyytikäinen LP, Nolte IM, Poveda A, Rauramaa R, Riaz M, Robino A, Sofer T, Takeuchi F, Tayo BO, van der Most PJ, Verweij N, Ware EB, Weiss S, Wen W, Yanek LR, Zhan Y, Amin N, Arking DE, Ballantyne C, Boerwinkle E, Brody JA, Broeckel U, Campbell A, Canouil M, Chai X, Chen YDI, Chen X, Chitrala KN, Concas MP, de Faire U, de Mutsert R, de Silva HJ, de Vries PS, Do A, Faul JD, Fisher V, Floyd JS, Forrester T, Friedlander Y, Girotto G, Gu CC, Hallmans G, Heikkinen S, Heng CK, Homuth G, Hunt S, Ikram MA, Jacobs DR, Kavousi M, Khor CC, Kilpeläinen TO, Koh WP, Komulainen P, Langefeld CD, Liang J, Liu K, Liu J, Lohman K, Mägi R, Manichaikul AW, McKenzie CA, Meitinger T, Milaneschi Y, Nauck M, Nelson CP, O’Connell JR, Palmer ND, Pereira AC, Perls T, Peters A, Polašek O, Raitakari OT, Rice K, Rice TK, Rich SS, Sabanayagam C, Schreiner PJ, Shu XO, Sidney S, Sims M, Smith JA, Starr JM, Strauch K, Tai ES, Taylor KD, Tsai MY, Uitterlinden AG, van Heemst D, Waldenberger M, Wang YX, Wei WB, Wilson G, Xuan D, Yao J, Yu C, Yuan JM, Zhao W, Becker DM, Bonnefond A, Bowden DW, Cooper RS, Deary IJ, Divers J, Esko T, Franks PW, Froguel P, Gieger C, Jonas JB, Kato N, Lakka TA, Leander K, Lehtimäki T, Magnusson PKE, North KE, Ntalla I, Penninx B, Samani NJ, Snieder H, Spedicati B, van der Harst P, Völzke H, Wagenknecht LE, Weir DR, Wojczynski MK, Wu T, Zheng W, Zhu X, Bouchard C, Chasman DI, Evans MK, Fox ER, Gudnason V, Hayward C, Horta BL, Kardia SLR, Krieger JE, Mook-Kanamori DO, Peyser PA, Province MM, Psaty BM, Rudan I, Sim X, Smith BH, van Dam RM, van Duijn CM, Wong TY, Arnett DK, Rao DC, Gauderman J, Liu CT, Morrison AC, Rotter JI, Fornage M. Gene-educational attainment interactions in a multi-population genome-wide meta-analysis identify novel lipid loci. Front Genet 2023; 14:1235337. [PMID: 38028628 PMCID: PMC10651736 DOI: 10.3389/fgene.2023.1235337] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/27/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction: Educational attainment, widely used in epidemiologic studies as a surrogate for socioeconomic status, is a predictor of cardiovascular health outcomes. Methods: A two-stage genome-wide meta-analysis of low-density lipoprotein cholesterol (LDL), high-density lipoprotein cholesterol (HDL), and triglyceride (TG) levels was performed while accounting for gene-educational attainment interactions in up to 226,315 individuals from five population groups. We considered two educational attainment variables: "Some College" (yes/no, for any education beyond high school) and "Graduated College" (yes/no, for completing a 4-year college degree). Genome-wide significant (p < 5 × 10-8) and suggestive (p < 1 × 10-6) variants were identified in Stage 1 (in up to 108,784 individuals) through genome-wide analysis, and those variants were followed up in Stage 2 studies (in up to 117,531 individuals). Results: In combined analysis of Stages 1 and 2, we identified 18 novel lipid loci (nine for LDL, seven for HDL, and two for TG) by two degree-of-freedom (2 DF) joint tests of main and interaction effects. Four loci showed significant interaction with educational attainment. Two loci were significant only in cross-population analyses. Several loci include genes with known or suggested roles in adipose (FOXP1, MBOAT4, SKP2, STIM1, STX4), brain (BRI3, FILIP1, FOXP1, LINC00290, LMTK2, MBOAT4, MYO6, SENP6, SRGAP3, STIM1, TMEM167A, TMEM30A), and liver (BRI3, FOXP1) biology, highlighting the potential importance of brain-adipose-liver communication in the regulation of lipid metabolism. An investigation of the potential druggability of genes in identified loci resulted in five gene targets shown to interact with drugs approved by the Food and Drug Administration, including genes with roles in adipose and brain tissue. Discussion: Genome-wide interaction analysis of educational attainment identified novel lipid loci not previously detected by analyses limited to main genetic effects.
Collapse
Affiliation(s)
- Lisa de las Fuentes
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, United States
| | - Karen L. Schwander
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, United States
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
| | - Michael R. Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Amy R. Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Thomas W. Winkler
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, United States
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - Patricia B. Munroe
- Clinical Pharmacology, Queen Mary University of London, London, United Kingdom
- National Institute for Health Research Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, London, United Kingdom
| | - Clint L. Miller
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA, United States
- Biochemistry and Molecular Genetics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA, United States
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, United States
| | - Hugo Aschard
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, United States
- Département de Génomes et Génétique, Institut Pasteur de Lille, Université de Lille, Lille, France
| | - Stella Aslibekyan
- School of Public Health, Epidemiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Traci M. Bartz
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, United States
- Department of Biostatistics, University of Washington, Seattle, WA, United States
| | - Lawrence F. Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Jin Fang Chai
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Ching-Yu Cheng
- Ocular Epidemiology, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, Medical School, Duke-National University of Singapore, Singapore, Singapore
| | - Rajkumar Dorajoo
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Mary F. Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
| | - Xiuqing Guo
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Los Angeles, CA, United States
| | - Fernando P. Hartwig
- Postgraduate Programme in Epidemiology, Faculty of Medicine, Federal University of Pelotas, Pelotas, RS, Brazil
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Andrea Horimoto
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of Sao Paulo Medical School, Sao Paulo, SP, Brazil
| | - Ivana Kolčić
- University of Split School of Medicine, Split, Croatia
- Algebra University College, Zagreb, Croatia
| | - Elise Lim
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States
| | - Yongmei Liu
- Division of Cardiology, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, United States
| | - Alisa K. Manning
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Jonathan Marten
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Solomon K. Musani
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Raymond Noordam
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Melissa A. Richard
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Paul M. Ridker
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Albert V. Smith
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, United States
- Icelandic Heart Association, Kopavogur, Iceland
| | - Dina Vojinovic
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, Netherlands
| | - Alan B. Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
- National Institutes of Health, Baltimore, MD, United States
| | - Maris Alver
- Estonian Genome Center, Insititute of Genomics, University of Tartu, Tartu, Estonia
| | - Mathilde Boissel
- European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille, France
- University of Lille, Lille University Hospital, Lille, France
| | - Kaare Christensen
- Unit of Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Barry I. Freedman
- Nephrology Division, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Chuan Gao
- Molecular Genetics and Genomics Program, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Franco Giulianini
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Sarah E. Harris
- Department of Psychology, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - Meian He
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang-Chi Hsu
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Brigitte Kühnel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Federica Laguzzi
- Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Xiaoyin Li
- Department of Population and Quantitative Health Sciences, Cleveland, OH, United States
- Department of Mathematics and Statistics, St. Cloud State University, St. Cloud, MN, United States
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, University of Tampere, Tampere, Finland
- Finnish Cardiovascular Research Center, University of Tampere, Tampere, Finland
| | - Ilja M. Nolte
- Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Alaitz Poveda
- Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Rainer Rauramaa
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Muhammad Riaz
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, United Kingdom
| | - Antonietta Robino
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - Tamar Sofer
- Biostatistics, Department of Medicine, Brigham and Women’s Hospital, Harvard University, Boston, MA, United States
| | - Fumihiko Takeuchi
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Bamidele O. Tayo
- Department of Public Health Sciences, Loyola University Chicago, Maywood, IL, United States
| | - Peter J. van der Most
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Niek Verweij
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Erin B. Ware
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, United States
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald and University of Greifswald, Greifswald, Germany
- German Center for Cardiovascular Research, Greifswald, Germany
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Lisa R. Yanek
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yiqiang Zhan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Dan E. Arking
- Department of Genetic Medicine, McKusick-Nathans Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Christie Ballantyne
- Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX, United States
- Houston Methodist Debakey Heart and Vascular Center, Houston, TX, United States
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, United States
| | - Jennifer A. Brody
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, United States
| | - Ulrich Broeckel
- Section on Genomic Pediatrics, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, United Kingdom
- Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, United Kingdom
| | - Mickaël Canouil
- European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille, France
- University of Lille, Lille University Hospital, Lille, France
| | - Xiaoran Chai
- Data Science Unit, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Yii-Der Ida Chen
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Los Angeles, CA, United States
| | - Xu Chen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Kumaraswamy Naidu Chitrala
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Maria Pina Concas
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - Ulf de Faire
- Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - H. Janaka de Silva
- Department of Medicine, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - Paul S. de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Ahn Do
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, United States
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
| | - Jessica D. Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, United States
| | - Virginia Fisher
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States
| | - James S. Floyd
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, United States
| | - Terrence Forrester
- Tropical Medicine Research Institute, University of the West Indies, Mona, Jamaica
| | - Yechiel Friedlander
- Braun School of Public Health, Hadassah Medical Center, Hebrew University, Jerusalem, Israel
| | - Giorgia Girotto
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - C. Charles Gu
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, United States
| | - Göran Hallmans
- Section for Nutritional Research, Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Sami Heikkinen
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Chew-Kiat Heng
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Khoo Teck Puat National University Children’s Medical Institute, National University Health System, Singapore, Singapore
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald and University of Greifswald, Greifswald, Germany
| | - Steven Hunt
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
- Department of Genetic Medicine, Weill Cornell Medicine in Qatar, Doha, Qatar
| | - M. Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - David R. Jacobs
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, United States
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Chiea Chuen Khor
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Tuomas O. Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Environmental Medicine and Public Health, The Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Woon-Puay Koh
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | | | - Carl D. Langefeld
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Jingjing Liang
- Department of Population and Quantitative Health Sciences, Cleveland, OH, United States
| | - Kiang Liu
- Epidemiology, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jianjun Liu
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Kurt Lohman
- Division of Cardiology, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, United States
| | - Reedik Mägi
- Estonian Genome Center, Insititute of Genomics, University of Tartu, Tartu, Estonia
| | - Ani W. Manichaikul
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA, United States
| | - Colin A. McKenzie
- Tropical Medicine Research Institute, University of the West Indies, Mona, Jamaica
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Human Genetics, Technische Universität München, Munich, Germany
| | | | - Matthias Nauck
- German Center for Cardiovascular Research, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Christopher P. Nelson
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, United Kingdom
| | - Jeffrey R. O’Connell
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, United States
- Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Nicholette D. Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Alexandre C. Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of Sao Paulo Medical School, Sao Paulo, SP, Brazil
| | - Thomas Perls
- Geriatrics Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Cardiovascular Research, Neuherberg, Germany
| | - Ozren Polašek
- University of Split School of Medicine, Split, Croatia
- Algebra University College, Zagreb, Croatia
| | - Olli T. Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Kenneth Rice
- Department of Biostatistics, University of Washington, Seattle, WA, United States
| | - Treva K. Rice
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, United States
| | - Stephen S. Rich
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA, United States
| | - Charumathi Sabanayagam
- Ocular Epidemiology, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, Medical School, Duke-National University of Singapore, Singapore, Singapore
| | - Pamela J. Schreiner
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, United States
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Stephen Sidney
- Division of Research, Kaiser Permanente of Northern California, Oakland, CA, United States
| | - Mario Sims
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Jennifer A. Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, United States
| | - John M. Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
- Alzheimer Scotland Dementia Research Centre, The University of Edinburgh, Edinburgh, United Kingdom
| | - Konstantin Strauch
- German Research Center for Environmental Health, Helmholtz Zentrum München, Institute of Genetic Epidemiology, Neuherberg, Germany
- Institute of Medical Informatics Biometry and Epidemiology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - E. Shyong Tai
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
- Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Kent D. Taylor
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Los Angeles, CA, United States
| | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, Minneapolis, MN, United States
| | - André G. Uitterlinden
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Diana van Heemst
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Ya-Xing Wang
- Beijing Ophthalmology and Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Institute of Ophthalmology, Capital Medical University, Beijing, China
| | - Wen-Bin Wei
- Beijing Ophthalmology and Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Institute of Ophthalmology, Capital Medical University, Beijing, China
| | - Gregory Wilson
- Jackson Heart Study Graduate Training Center, School of Public, Jackson State University, Jackson, MS, United States
| | - Deng Xuan
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States
| | - Jie Yao
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Los Angeles, CA, United States
| | - Caizheng Yu
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Min Yuan
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
- Division of Cancer Control and Population Sciences, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Diane M. Becker
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Amélie Bonnefond
- European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille, France
- University of Lille, Lille University Hospital, Lille, France
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Donald W. Bowden
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Richard S. Cooper
- Department of Public Health Sciences, Loyola University Chicago, Maywood, IL, United States
| | - Ian J. Deary
- Department of Psychology, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - Jasmin Divers
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Tõnu Esko
- Estonian Genome Center, Insititute of Genomics, University of Tartu, Tartu, Estonia
- Broad Institute, Massachusetts Institute of Technology and Harvard University, Boston, MA, United States
| | - Paul W. Franks
- Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Skåne University Hospital, Lund University, Malmö, Sweden
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
- Department of Nutrition, Harvard Chan School of Public Health, Boston, MA, United States
| | - Philippe Froguel
- European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille, France
- University of Lille, Lille University Hospital, Lille, France
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Jost B. Jonas
- Beijing Ophthalmology and Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Institute of Ophthalmology, Capital Medical University, Beijing, China
- Department of Ophthalmology, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
- Institute of Molecular and Clinical Ophthalmology, Basel, Switzerland
| | - Norihiro Kato
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Timo A. Lakka
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Karin Leander
- Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Terho Lehtimäki
- Department of Clinical Chemistry, University of Tampere, Tampere, Finland
- Finnish Cardiovascular Research Center, University of Tampere, Tampere, Finland
| | - Patrik K. E. Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Kari E. North
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ioanna Ntalla
- Clinical Pharmacology, Queen Mary University of London, London, United Kingdom
- Celgene, Bristol Myers Squibb, Mississauga, ON, Canada
| | | | - Nilesh J. Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, United Kingdom
| | - Harold Snieder
- Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Beatrice Spedicati
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Pim van der Harst
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Henry Völzke
- German Center for Cardiovascular Research, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Lynne E. Wagenknecht
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - David R. Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, United States
| | - Mary K. Wojczynski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
| | - Tangchun Wu
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Xiaofeng Zhu
- Department of Population and Quantitative Health Sciences, Cleveland, OH, United States
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Daniel I. Chasman
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Michele K. Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
- National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
| | - Ervin R. Fox
- Division of Cardiology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Bernardo L. Horta
- Postgraduate Programme in Epidemiology, Faculty of Medicine, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Sharon L. R. Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Jose Eduardo Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of Sao Paulo Medical School, Sao Paulo, SP, Brazil
| | - Dennis O. Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, Netherlands
| | - Patricia A. Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Michael M. Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, United States
- Department of Epidemiology, University of Washington, Seattle, WA, United States
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, United States
| | - Igor Rudan
- Centre for Global Health, The Usher Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Blair H. Smith
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Rob M. van Dam
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Department of Exercise and Nutrition Sciences, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| | - Cornelia M. van Duijn
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Tien Yin Wong
- Ocular Epidemiology, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, Medical School, Duke-National University of Singapore, Singapore, Singapore
| | - Donna K. Arnett
- College of Public Health, Dean’s Office, University of Kentucky, Lexington, KY, United States
| | - Dabeeru C. Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, United States
| | - James Gauderman
- Division of Biostatistics, Population and Public Health Sciences, University of Southern California, Los Angeles, CA, United States
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States
| | - Alanna C. Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jerome I. Rotter
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Los Angeles, CA, United States
| | - Myriam Fornage
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
36
|
Carr KD, Weiner SP, Vasquez C, Schmidt AM. Involvement of the Receptor for Advanced Glycation End Products (RAGE) in high fat-high sugar diet-induced anhedonia in rats. Physiol Behav 2023; 271:114337. [PMID: 37625475 PMCID: PMC10592025 DOI: 10.1016/j.physbeh.2023.114337] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/08/2023] [Accepted: 08/21/2023] [Indexed: 08/27/2023]
Abstract
Clinical and basic science investigation indicates a link between insulin resistance and anhedonia. Previous results of this laboratory point to impaired nucleus accumbens (NAc) insulin signaling as an underpinning of diet-induced anhedonia, based on use of a glucose lick microstructure assay. The present study evaluated whether advanced glycation end products (AGEs) and their receptor (RAGE), known to mediate obesogenic diet-induced inflammation and pathological metabolic conditions, are involved in this behavioral change. Six weeks maintenance of male and female rats on a high fat-high sugar liquid diet (chocolate Ensure) increased body weight gain, and markedly increased circulating insulin and leptin, but induced anhedonia (decreased first minute lick rate and lick burst size) in males only. In these subjects, anhedonia correlated with plasma concentrations of insulin. Although the diet did not alter plasma or NAc AGEs, or the expression of RAGE in the NAc, marginally significant correlations were seen between anhedonia and plasma content of several AGEs and NAc RAGE. Importantly, a small molecule RAGE antagonist, RAGE229, administered twice daily by oral gavage, prevented diet-induced anhedonia. This beneficial effect was associated with improved adipose function, reflected in the adiponectin/leptin ratio, and increased pCREB/total CREB in the NAc, and a shift in the pCREB correlation with pThr34-DARPP-32 from near-zero to strongly positive, such that both phospho-proteins correlated with the rescued hedonic response. This set of findings suggests that the receptor/signaling pathway and cell type underlying the RAGE229-mediated increase in pCREB may mediate anhedonia and its prevention. The possible role of adipose tissue as a locus of diet-induced RAGE signaling, and source of circulating factors that target NAc to modify hedonic reactivity are discussed.
Collapse
Affiliation(s)
- Kenneth D Carr
- Departments of Psychiatry, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States; Departments of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States.
| | - Sydney P Weiner
- Departments of Psychiatry, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
| | - Carolina Vasquez
- Departments of Psychiatry, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States; Departments of Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
| | - Ann Marie Schmidt
- Departments of Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, United States
| |
Collapse
|
37
|
Huang F, Marungruang N, Martinsson I, Camprubí Ferrer L, Nguyen TD, Gondo TF, Karlsson EN, Deierborg T, Öste R, Heyman-Lindén L. A mixture of Nordic berries improves cognitive function, metabolic function and alters the gut microbiota in C57Bl/6J male mice. Front Nutr 2023; 10:1257472. [PMID: 37854349 PMCID: PMC10580983 DOI: 10.3389/fnut.2023.1257472] [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: 07/12/2023] [Accepted: 09/18/2023] [Indexed: 10/20/2023] Open
Abstract
Our diets greatly influence our health. Multiple lines of research highlight the beneficial properties of eating berries and fruits. In this study, a berry mixture of Nordic berries previously identified as having the potential to improve memory was supplemented to young C57Bl/6J male mice to investigate effects on cognition function, metabolic health, markers of neuroinflammation, and gut microbiota composition. C57Bl/6J male mice at the age of 8 weeks were given standard chow, a high-fat diet (HF, 60%E fat), or a high-fat diet supplemented with freeze-dried powder (20% dwb) of a mixture of Nordic berries and red grape juice (HF + Berry) for 18 weeks (n = 12 animals/diet group). The results show that supplementation with the berry mixture may have beneficial effects on spatial memory, as seen by enhanced performance in the T-maze and Barnes maze compared to the mice receiving the high-fat diet without berries. Additionally, berry intake may aid in counteracting high-fat diet induced weight gain and could influence neuroinflammatory status as suggested by the increased levels of the inflammation modifying IL-10 cytokine in hippocampal extracts from berry supplemented mice. Furthermore, the 4.5-month feeding with diet containing berries resulted in significant changes in cecal microbiota composition. Analysis of cecal bacterial 16S rRNA revealed that the chow group had significantly higher microbial diversity, as measured by the Shannon diversity index and total operational taxonomic unit richness, than the HF group. The HF diet supplemented with berries resulted in a strong trend of higher total OTU richness and significantly increased the relative abundance of Akkermansia muciniphila, which has been linked to protective effects on cognitive decline. In conclusion, the results of this study suggest that intake of a Nordic berry mixture is a valuable strategy for maintaining and improving cognitive function, to be further evaluated in clinical trials.
Collapse
Affiliation(s)
- Fang Huang
- Division of Biotechnology, Department of Chemistry, Lund University, Lund, Sweden
- Aventure AB, Lund, Sweden
| | | | - Isak Martinsson
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Lluís Camprubí Ferrer
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Thao Duy Nguyen
- Department of Food Technology, Engineering and Nutrition, Lund University, Lund, Sweden
| | - Thamani Freedom Gondo
- Department of Chemistry, Centre for Analysis and Synthesis, Lund University, Lund, Sweden
| | | | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Lovisa Heyman-Lindén
- Berry Lab AB, Lund, Sweden
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
38
|
Ullah R, Shen Y, Zhou YD, Fu J. Perinatal metabolic inflammation in the hypothalamus impairs the development of homeostatic feeding circuitry. Metabolism 2023; 147:155677. [PMID: 37543245 DOI: 10.1016/j.metabol.2023.155677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/14/2023] [Accepted: 08/01/2023] [Indexed: 08/07/2023]
Abstract
Over the past few decades, there has been a global increase in childhood obesity. This rise in childhood obesity contributes to the susceptibility of impaired metabolism during both childhood and adulthood. The hypothalamus, specifically the arcuate nucleus (ARC), houses crucial neurons involved in regulating homeostatic feeding. These neurons include proopiomelanocortin (POMC) and agouti-related peptide (AGRP) secreting neurons. They play a vital role in sensing nutrients and metabolic hormones like insulin, leptin, and ghrelin. The neurogenesis of AGRP and POMC neurons completes at birth; however, axon development and synapse formation occur during the postnatal stages in rodents. Insulin, leptin, and ghrelin are the essential regulators of POMC and AGRP neurons. Maternal obesity and postnatal overfeeding or a high-fat diet (HFD) feeding cause metabolic inflammation, disrupted signaling of metabolic hormones, netrin-1, and neurogenic factors, neonatal obesity, and defective neuronal development in animal models; however, the mechanism is unclear. Within the hypothalamus and other brain areas, there exists a wide range of interconnected neuronal populations that regulate various aspects of feeding. However, this review aims to discuss how perinatal metabolic inflammation influences the development of POMC and AGRP neurons within the hypothalamus.
Collapse
Affiliation(s)
- Rahim Ullah
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, 310052, China; Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China
| | - Yi Shen
- Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China.
| | - Yu-Dong Zhou
- Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China.
| | - Junfen Fu
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, 310052, China.
| |
Collapse
|
39
|
Huguet G, Puig-Parnau I, Serrano JCE, Martin-Gari M, Rodríguez-Palmero M, Moreno-Muñoz JA, Tibau J, Kádár E. Hippocampal neurogenesis and Arc expression are enhanced in high-fat fed prepubertal female pigs by a diet including omega-3 fatty acids and Bifidobacterium breve CECT8242. Eur J Nutr 2023; 62:2463-2473. [PMID: 37148357 PMCID: PMC10421764 DOI: 10.1007/s00394-023-03165-1] [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: 01/12/2023] [Accepted: 04/21/2023] [Indexed: 05/08/2023]
Abstract
PURPOSE Obesity during childhood has become a pandemic disease, mainly caused by a diet rich in sugars and fatty acids. Among other negative effects, these diets can induce cognitive impairment and reduce neuroplasticity. It is well known that omega-3 and probiotics have a beneficial impact on health and cognition, and we have hypothesized that a diet enriched with Bifidobacterium breve and omega-3 could potentiate neuroplasticity in prepubertal pigs on a high-fat diet. METHODS Young female piglets were fed during 10 weeks with: standard diet (T1), high-fat (HF) diet (T2), HF diet including B. breve CECT8242 (T3) and HF diet including the probiotic and omega-3 fatty acids (T4). Using hippocampal sections, we analyzed by immunocytochemistry the levels of doublecortin (DCX) to study neurogenesis, and activity-regulated cytoskeleton-associated protein (Arc) as a synaptic plasticity related protein. RESULTS No effect of T2 or T3 was observed, whereas T4 increased both DCX+ cells and Arc expression. Therefore, a diet enriched with supplements of B. breve and omega-3 increases neurogenesis and synaptic plasticity in prepubertal females on a HF diet from nine weeks of age to sexual maturity. Furthermore, the analysis of serum cholesterol and HDL indicate that neurogenesis was related to lipidic demand in piglets fed with control or HF diets, but the neurogenic effect induced by the T4 diet was exerted by mechanisms independent of this lipidic demand. CONCLUSION Our results show that the T4 dietary treatment is effective in potentiating neural plasticity in the dorsal hippocampus of prepubertal females on a HF diet.
Collapse
Affiliation(s)
- Gemma Huguet
- Department of Biology, Universitat de Girona, Girona, Spain
| | | | - Jose C. E. Serrano
- IRBLleida-Universitat de Lleida, Avda Rovira Roure 80, 25196 Lleida, Spain
| | | | | | | | - Joan Tibau
- Animal Science-Institut de Recerca i Tecnologia Agroalimentàries, IRTA-Monells, 17121 Monells, Spain
| | - Elisabet Kádár
- Department of Biology, Universitat de Girona, Girona, Spain
| |
Collapse
|
40
|
Mohammadi S, Lotfi K, Mokhtari E, Hajhashemy Z, Heidari Z, Saneei P. Association between Mediterranean dietary pattern with sleep duration, sleep quality and brain derived neurotrophic factor (BDNF) in Iranian adults. Sci Rep 2023; 13:13493. [PMID: 37596403 PMCID: PMC10439234 DOI: 10.1038/s41598-023-40625-4] [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: 09/25/2022] [Accepted: 08/14/2023] [Indexed: 08/20/2023] Open
Abstract
Data on the association between Mediterranean diet, sleep and brain-derived neurotrophic factor (BDNF) were limited in Middle Eastern populations. We examined the association between Mediterranean dietary pattern with sleep quality/quantity, and serum BDNF in Iranian adults. This cross-sectional study was performed among 535 middle-aged adults (54% men), selected by multistage cluster random sampling method. The Pittsburgh sleep quality index and a validated food frequency questionnaire were used to assess sleep quality, sleep quantity, and Mediterranean diet score (MDS). Twelve-hour fasting blood samples were taken to evaluate serum BDNF values. Participants in the highest tertile of MDS, in comparison to those in the lowest tertile, had lower odds of having short sleep (OR = 0.44, 95%CI: 0.21-0.91) and poor sleep quality (OR = 0.48, 95%CI: 0.22-0.96), after adjustment for potential confounders. Among specific domains of sleep quality, lower odds of subjective sleep quality, sleep latency, and daytime dysfunction were associated with increased MDS. Higher adherence to MDS among individuals with overweight or obesity reduced the odds of having short sleep; this relation was not seen among individuals with normal weight. In contrast, the association between sleep quality and the MDS was significant in individuals with normal weight, but not those with overweight or obesity. Participants with higher adherence to MDS had lower odds for low BDNF values; however, this relation was not statistically significant. Overall, Iranian adults with a higher adherence to MDS had considerably lower odds of having short sleep and poor sleep quality. BDNF would not be an intermediate molecule for this connection.
Collapse
Affiliation(s)
- Sobhan Mohammadi
- Department of Community Nutrition, School of Nutrition and Food Science, Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, PO Box 81745-151, Isfahan, Iran
| | - Keyhan Lotfi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Mokhtari
- Department of Community Nutrition, School of Nutrition and Food Science, Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, PO Box 81745-151, Isfahan, Iran
| | - Zahra Hajhashemy
- Department of Community Nutrition, School of Nutrition and Food Science, Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, PO Box 81745-151, Isfahan, Iran
| | - Zahra Heidari
- Department of Biostatistics and Epidemiology, School of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parvane Saneei
- Department of Community Nutrition, School of Nutrition and Food Science, Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, PO Box 81745-151, Isfahan, Iran.
| |
Collapse
|
41
|
Jafari RS, Behrouz V. Nordic diet and its benefits in neurological function: a systematic review of observational and intervention studies. Front Nutr 2023; 10:1215358. [PMID: 37645628 PMCID: PMC10461010 DOI: 10.3389/fnut.2023.1215358] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/31/2023] [Indexed: 08/31/2023] Open
Abstract
Introduction Neurological disorders have been considered the major contributors to global long-term disability and lower quality of life. Lifestyle factors, such as dietary patterns, are increasingly recognized as important determinants of neurological function. Some dietary behaviors, such as Nordic diet (ND) were likely to have protective effects on brain function. However, an understanding of the effectiveness of the ND pattern to improve neurological function and brain health is not fully understood. We review the current evidence that supports the ND pattern in various aspects of neurological function and addresses both proven and less established mechanisms of action based on its food ingredients and biochemical compounds. Methods In this systematic review, PubMed, Web of Science, and Scopus databases were searched from inception to February 2023. Observational and intervention studies were included. Results Of the 627 screened studies, 5 observational studies (including three cohorts and two cross-sectional studies) and 3 intervention studies investigating the association between ND and neurological function. Observational studies investigated the association of ND with the following neurological functions: cognition, stroke, and neuropsychological function. Intervention studies investigated the effects of ND on cognition and depression. Discussion Despite the limited literature on ND and its association with neurological function, several aspects of ND may lead to some health benefits suggesting neuroprotective effects. The current state of knowledge attributes the possible effects of characteristic components of the ND to its antioxidant, anti-inflammatory, lipid-lowering, gut-brain-axis modulating, and ligand activities in cell signaling pathways. Based on existing evidence, the ND may be considered a recommended dietary approach for the improvement of neurological function and brain health. Systematic review registration [https://www.crd.york.ac.uk/prospero/], identifier [CRD2023451117].
Collapse
Affiliation(s)
| | - Vahideh Behrouz
- Department of Nutrition, Faculty of Public Health, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
42
|
Doroszkiewicz J, Mroczko J, Rutkowski P, Mroczko B. Molecular Aspects of a Diet as a New Pathway in the Prevention and Treatment of Alzheimer's Disease. Int J Mol Sci 2023; 24:10751. [PMID: 37445928 PMCID: PMC10341644 DOI: 10.3390/ijms241310751] [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: 05/22/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Alzheimer's disease is the most common cause of dementia in the world. Lack of an established pathology makes it difficult to develop suitable approaches and treatment for the disease. Besides known hallmarks, including amyloid β peptides cumulating in plaques and hyperphosphorylated tau forming NFTs, inflammation also plays an important role, with known connections to the diet. In AD, adhering to reasonable nutrition according to age-related principles is recommended. The diet should be high in neuroprotective foods, such as polyunsaturated fatty acids, antioxidants, and B vitamins. In addition, foods capable of rising BDNF should be considered because of the known profitable results of this molecule in AD. Adhering to beneficial diets might result in improvements in memory, cognition, and biomarkers and might even reduce the risk of developing AD. In this review, we discuss the effects of various diets, foods, and nutrients on brain health and possible connections to Alzheimer's disease.
Collapse
Affiliation(s)
- Julia Doroszkiewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-089 Bialystok, Poland; (J.M.); (B.M.)
| | - Jan Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-089 Bialystok, Poland; (J.M.); (B.M.)
| | | | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-089 Bialystok, Poland; (J.M.); (B.M.)
- Department of Biochemical Diagnostics, Medical University of Białystok, 15-089 Bialystok, Poland
| |
Collapse
|
43
|
Wang S, Zeng F, Ma Y, Yu J, Xiang C, Feng X, Wang S, Wang J, Zhao S, Zhu X. Strontium Attenuates Hippocampal Damage via Suppressing Neuroinflammation in High-Fat Diet-Induced NAFLD Mice. Int J Mol Sci 2023; 24:10248. [PMID: 37373395 DOI: 10.3390/ijms241210248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) leads to hippocampal damage and causes a variety of physiopathological responses, including the induction of endoplasmic reticulum stress (ERS), neuroinflammation, and alterations in synaptic plasticity. As an important trace element, strontium (Sr) has been reported to have antioxidant effects, to have anti-inflammatory effects, and to cause the inhibition of adipogenesis. The present study was undertaken to investigate the protective effects of Sr on hippocampal damage in NAFLD mice in order to elucidate the underlying mechanism of Sr in NAFLD. The mouse model of NAFLD was established by feeding mice a high-fat diet (HFD), and the mice were treated with Sr. In the NAFLD mice, we found that treatment with Sr significantly increased the density of c-Fos+ cells in the hippocampus and inhibited the expression of caspase-3 by suppressing ERS. Surprisingly, the induction of neuroinflammation and the increased expression of inflammatory cytokines in the hippocampus following an HFD were attenuated by Sr treatment. Sr significantly attenuated the activation of microglia and astrocytes induced by an HFD. The expression of phospho-p38, ERK, and NF-κB was consistently significantly increased in the HFD group, and treatment with Sr decreased their expression. Moreover, Sr prevented HFD-induced damage to the ultra-structural synaptic architecture. This study implies that Sr has beneficial effects on repairing the damage to the hippocampus induced by an HFD, revealing that Sr could be a potential candidate for protection from neural damage caused by NAFLD.
Collapse
Affiliation(s)
- Shuai Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Fangyuan Zeng
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Yue Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Jiaojiao Yu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Chenyao Xiang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Xiao Feng
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Songlin Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Jianguo Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Xiaoyan Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| |
Collapse
|
44
|
Ab-Hamid N, Omar N, Ismail CAN, Long I. Diabetes and cognitive decline: Challenges and future direction. World J Diabetes 2023; 14:795-807. [PMID: 37383592 PMCID: PMC10294066 DOI: 10.4239/wjd.v14.i6.795] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/07/2023] [Accepted: 05/06/2023] [Indexed: 06/14/2023] Open
Abstract
There is growing evidence that diabetes can induce cognitive decline and dementia. It is a slow, progressive cognitive decline that can occur in any age group, but is seen more frequently in older individuals. Symptoms related to cognitive decline are worsened by chronic metabolic syndrome. Animal models are frequently utilized to elucidate the mechanisms of cognitive decline in diabetes and to assess potential drugs for therapy and prevention. This review addresses the common factors and pathophysiology involved in diabetes-related cognitive decline and outlines the various animal models used to study this condition.
Collapse
Affiliation(s)
- Norhamidar Ab-Hamid
- Biomedicine program, School of Health Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Kelantan, Malaysia
| | - Norsuhana Omar
- Department of Physiology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Kelantan, Malaysia
| | - Che Aishah Nazariah Ismail
- Department of Physiology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Kelantan, Malaysia
| | - Idris Long
- Biomedicine program, School of Health Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Kelantan, Malaysia
| |
Collapse
|
45
|
Song J. Amygdala activity and amygdala-hippocampus connectivity: Metabolic diseases, dementia, and neuropsychiatric issues. Biomed Pharmacother 2023; 162:114647. [PMID: 37011482 DOI: 10.1016/j.biopha.2023.114647] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/04/2023] Open
Abstract
With rapid aging of the population worldwide, the number of people with dementia is dramatically increasing. Some studies have emphasized that metabolic syndrome, which includes obesity and diabetes, leads to increased risks of dementia and cognitive decline. Factors such as insulin resistance, hyperglycemia, high blood pressure, dyslipidemia, and central obesity in metabolic syndrome are associated with synaptic failure, neuroinflammation, and imbalanced neurotransmitter levels, leading to the progression of dementia. Due to the positive correlation between diabetes and dementia, some studies have called it "type 3 diabetes". Recently, the number of patients with cognitive decline due to metabolic imbalances has considerably increased. In addition, recent studies have reported that neuropsychiatric issues such as anxiety, depressive behavior, and impaired attention are common factors in patients with metabolic disease and those with dementia. In the central nervous system (CNS), the amygdala is a central region that regulates emotional memory, mood disorders, anxiety, attention, and cognitive function. The connectivity of the amygdala with other brain regions, such as the hippocampus, and the activity of the amygdala contribute to diverse neuropathological and neuropsychiatric issues. Thus, this review summarizes the significant consequences of the critical roles of amygdala connectivity in both metabolic syndromes and dementia. Further studies on amygdala function in metabolic imbalance-related dementia are needed to treat neuropsychiatric problems in patients with this type of dementia.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea.
| |
Collapse
|
46
|
Maryam K, Ali H. Aerobic and resistance exercises affect the BDNF/TrkB signaling pathway, and hippocampal neuron density of high-fat diet-induced obese elderly rats. Physiol Behav 2023; 264:114140. [PMID: 36870384 DOI: 10.1016/j.physbeh.2023.114140] [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: 11/21/2022] [Revised: 02/05/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
Obesity, caused by a high-fat diet (HFD) in elderly, is a risk factor for insulin resistance and a precursor to diabetes and can lead to impaired cognitive function. Physical exercise has positive effects on decrease obesity and improvement brain function. We investigated which type of aerobic (AE) or resistance (RE) exercise can be more effective in reducing HFD-induced cognitive dysfunction in obese elderly rats. 48 male Wistar rats (19-monthold) were divided into six groups: Healthy control (CON), CON+AE, CON+RE, HFD, HFD+AE, and HFD+RE. Obesity was induced by 5 months of HFD feeding in older rats. After obesity confirmation, RT (with a range of 50% to 100%1RM/3 days/week) and AE (running at 8-m/min for 15-min to 26-m/min for 60-min /5 days/week) was performed for 12-weeks. Morris water maze Test was used to evaluate cognitive performance. All data were analyzed using two-way statistical test of variance. The results showed that obesity had a negative effect on glycemic index, increased inflammation, decreased antioxidant levels, decreased BDNF/TrkB and decreased nerve density in hippocampal tissue. The Morris water maze results clearly showed cognitive impairment in the obesity group. But 12 weeks after AE and RE, all the measured variables were on the improvement path, and in general, no difference was observed between the two exercise methods. Two mods of exercise (AE and RE) may be having same effects on nerve cell density, inflammatory, antioxidant and functional status of hippocampus of obese rats. Each of the AE and RE can create beneficial effects on the cognitive function of the elderly.
Collapse
Affiliation(s)
- Keshvari Maryam
- Faculty of Sport Sciences, Exercise Physiology Department, Bu-Ali Sina University, Hamadan, Iran
| | - Heidarianpour Ali
- Faculty of Sport Sciences, Exercise Physiology Department, Bu-Ali Sina University, Hamadan, Iran.
| |
Collapse
|
47
|
Cai Y, Liu P, Zhou X, Yuan J, Chen Q. Probiotics therapy show significant improvement in obesity and neurobehavioral disorders symptoms. Front Cell Infect Microbiol 2023; 13:1178399. [PMID: 37249983 PMCID: PMC10213414 DOI: 10.3389/fcimb.2023.1178399] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Obesity is a complex metabolic disease, with cognitive impairment being an essential complication. Gut microbiota differs markedly between individuals with and without obesity. The microbial-gut-brain axis is an important pathway through which metabolic factors, such as obesity, affect the brain. Probiotics have been shown to alleviate symptoms associated with obesity and neurobehavioral disorders. In this review, we evaluated previously published studies on the effectiveness of probiotic interventions in reducing cognitive impairment, depression, and anxiety associated with obesity or a high-fat diet. Most of the probiotics studied have beneficial health effects on obesity-induced cognitive impairment and anxiety. They positively affect immune regulation, the hypothalamic-pituitary-adrenal axis, hippocampal function, intestinal mucosa protection, and glucolipid metabolism regulation. Probiotics can influence changes in the composition of the gut microbiota and the ratio between various flora. However, probiotics should be used with caution, particularly in healthy individuals. Future research should further explore the mechanisms underlying the gut-brain axis, obesity, and cognitive function while overcoming the significant variation in study design and high risk of bias in the current evidence.
Collapse
|
48
|
Li ZA, Samara A, Ray MK, Rutlin J, Raji CA, Shimony JS, Sun P, Song SK, Hershey T, Eisenstein SA. Childhood obesity is linked to putative neuroinflammation in brain white matter, hypothalamus, and striatum. Cereb Cortex Commun 2023; 4:tgad007. [PMID: 37207193 PMCID: PMC10191798 DOI: 10.1093/texcom/tgad007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/19/2023] [Accepted: 04/22/2023] [Indexed: 05/21/2023] Open
Abstract
Neuroinflammation is both a consequence and driver of overfeeding and weight gain in rodent obesity models. Advances in magnetic resonance imaging (MRI) enable investigations of brain microstructure that suggests neuroinflammation in human obesity. To assess the convergent validity across MRI techniques and extend previous findings, we used diffusion basis spectrum imaging (DBSI) to characterize obesity-associated alterations in brain microstructure in 601 children (age 9-11 years) from the Adolescent Brain Cognitive DevelopmentSM Study. Compared with children with normal-weight, greater DBSI restricted fraction (RF), reflecting neuroinflammation-related cellularity, was seen in widespread white matter in children with overweight and obesity. Greater DBSI-RF in hypothalamus, caudate nucleus, putamen, and, in particular, nucleus accumbens, correlated with higher baseline body mass index and related anthropometrics. Comparable findings were seen in the striatum with a previously reported restriction spectrum imaging (RSI) model. Gain in waist circumference over 1 and 2 years related, at nominal significance, to greater baseline RSI-assessed restricted diffusion in nucleus accumbens and caudate nucleus, and DBSI-RF in hypothalamus, respectively. Here we demonstrate that childhood obesity is associated with microstructural alterations in white matter, hypothalamus, and striatum. Our results also support the reproducibility, across MRI methods, of findings of obesity-related putative neuroinflammation in children.
Collapse
Affiliation(s)
- Zhaolong Adrian Li
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, MO 63130, United States
| | - Amjad Samara
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110 United States
| | - Mary Katherine Ray
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
| | - Jerrel Rutlin
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
| | - Cyrus A Raji
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110 United States
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
| | - Joshua S Shimony
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
| | - Peng Sun
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Sheng-Kwei Song
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
| | - Tamara Hershey
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, MO 63130, United States
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110 United States
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
| | - Sarah A Eisenstein
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
| |
Collapse
|
49
|
Azbazdar Y, Poyraz YK, Ozalp O, Nazli D, Ipekgil D, Cucun G, Ozhan G. High-fat diet feeding triggers a regenerative response in the adult zebrafish brain. Mol Neurobiol 2023; 60:2486-2506. [PMID: 36670270 DOI: 10.1007/s12035-023-03210-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/04/2023] [Indexed: 01/22/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) includes a range of liver conditions ranging from excess fat accumulation to liver failure. NAFLD is strongly associated with high-fat diet (HFD) consumption that constitutes a metabolic risk factor. While HFD has been elucidated concerning its several systemic effects, there is little information about its influence on the brain at the molecular level. Here, by using a high-fat diet (HFD)-feeding of adult zebrafish, we first reveal that excess fat uptake results in weight gain and fatty liver. Prolonged exposure to HFD induces a significant increase in the expression of pro-inflammation, apoptosis, and proliferation markers in the liver and brain tissues. Immunofluorescence analyses of the brain tissues disclose stimulation of apoptosis and widespread activation of glial cell response. Moreover, glial activation is accompanied by an initial decrease in the number of neurons and their subsequent replacement in the olfactory bulb and the telencephalon. Long-term consumption of HFD causes activation of Wnt/β-catenin signaling in the brain tissues. Finally, fish fed an HFD induces anxiety, and aggressiveness and increases locomotor activity. Thus, HFD feeding leads to a non-traumatic brain injury and stimulates a regenerative response. The activation mechanisms of a regeneration response in the brain can be exploited to fight obesity and recover from non-traumatic injuries.
Collapse
Affiliation(s)
- Yagmur Azbazdar
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, 35340, Izmir, Turkey
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, CA, 90095-1662, USA
| | - Yusuf Kaan Poyraz
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, 35340, Izmir, Turkey
| | - Ozgun Ozalp
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, 35340, Izmir, Turkey
- Department of Molecular Life Sciences, University of Zurich, CH-8057, Zurich, Switzerland
| | - Dilek Nazli
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, 35340, Izmir, Turkey
| | - Dogac Ipekgil
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, 35340, Izmir, Turkey
| | - Gokhan Cucun
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, 35340, Izmir, Turkey
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), 3640 76021, Karlsruhe, Postfach, Germany
| | - Gunes Ozhan
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, 35340, Izmir, Turkey.
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, 35340, Izmir, Turkey.
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, 35430, Izmir, Turkey.
| |
Collapse
|
50
|
Walk AM, Cannavale CN, Keye SA, Rosok L, Edwards C, Khan N. Weight status impacts children's incidental statistical learning. Int J Psychophysiol 2023; 187:34-42. [PMID: 36796729 DOI: 10.1016/j.ijpsycho.2023.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023]
Abstract
The expanding literature investigating the cognitive effects of childhood weight status has not included examinations of incidental statistical learning, the process by which children unintentionally acquire knowledge about patterns in their environments, despite evidence that it underlies many higher-level information processing capabilities. In the present study, we measured event-related potentials (ERPs) while school-aged participants completed a variation of an oddball task in which stimuli predicted the appearance of a target. Children were asked to respond to the target but were not given any information about the existence of predictive dependencies. We found that children with a healthy weight status had larger P3 amplitudes in response to the predictors that were most meaningful in completing the task, a finding that may suggest optimized learning mechanisms influenced by weight status. These findings offer an important first step to understanding how healthy lifestyle factors may influence incidental statistical learning.
Collapse
Affiliation(s)
- Anne M Walk
- Eastern Illinois University, Department of Psychology, United States of America
| | - Corinne N Cannavale
- University of Illinois at Urbana-Champaign, Department of Kinesiology & Community Health, United States of America
| | - Shelby A Keye
- University of Illinois at Urbana-Champaign, Department of Kinesiology & Community Health, United States of America
| | - Laura Rosok
- University of Illinois at Urbana-Champaign, Neuroscience Program, United States of America
| | - Caitlyn Edwards
- University of Illinois at Urbana-Champaign, Division of Nutritional Sciences, United States of America
| | - Naiman Khan
- University of Illinois at Urbana-Champaign, Department of Kinesiology & Community Health, United States of America; University of Illinois at Urbana-Champaign, Division of Nutritional Sciences, United States of America; University of Illinois at Urbana-Champaign, Neuroscience Program, United States of America; University of Illinois at Urbana-Champaign, Beckman Institute of Advanced Science and Technology, United States of America.
| |
Collapse
|