1
|
Al Jaberi FM, Alzarzour R, Dewa A, Muhamad A, Zakaria F. Metabolic clues to memory loss: High-fat diets and brain-adipose crosstalk in zebrafish. Behav Brain Res 2025; 486:115559. [PMID: 40164316 DOI: 10.1016/j.bbr.2025.115559] [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: 02/05/2025] [Revised: 03/27/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Obesity is a growing public health concern that significantly impacts cognitive functions, including memory. This research explores how a high-fat diet affects short-term memory, employing the novel object recognition (NOR) test and NMR-based metabolomics to elucidate metabolic alterations in the brain and adipose tissue. The zebrafish were divided into two groups: one receiving a standard diet (SD) and the other a high-fat diet (HFD). Body mass index (BMI) was assessed every two weeks for a period of eight weeks. The NOR test was used to determine the discrimination index (DI) for evaluating the short-term memory of the SD and HFD groups. NMR spectroscopy was employed to investigate the metabolites in brain and adipose tissues, and multivariate data analysis was conducted to discover significant metabolic alterations. The high-fat diet (HFD) resulted in a significant increase in body mass index (BMI) (p < 0.0001) compared to the standard diet (SD) group from week 4 to week 8. A significant reduction in the discrimination index (24.95 %) in the HFD group against the SD group suggests a decline in memory performance among HFD subjects. NMR-based metabolomics of adipose tissue revealed that linoleic acid and caprylic acid were consistently found to exhibit increased levels in the HFD group across all assessments, whereas lauric acid, ALA, EPA, and DHA were consistently present at elevated levels in the adipose tissue of the SD group. NMR-based metabolomics of the brain identified GABA, taurine, and histamine as the key metabolites distinguishing the HFD from the SD group in female zebrafish. For male zebrafish brains, taurine, phenylalanine, and tryptophan were identified as the most significant metabolites for differentiating between HFD and SD. These metabolites demonstrated a notable decrease in the HFD group relative to the SD group. The results of this study align with those of previously reported studies in rodents and humans, indicating that memory impairment associated with obesity may stem from neuroinflammation and changes in synaptic plasticity. This research provides insights into the molecular changes in adipose tissue and the brain that occur when individuals receive a high-fat diet (HFD), which may enhance our understanding of the link between obesity and memory impairment, ultimately leading to a better comprehension of the disease.
Collapse
Affiliation(s)
- Farah Mejbel Al Jaberi
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia; Pharmaceutical Chemistry Department, Faculty of Pharmacy, University of Misan, Amarah, Maysan, Iraq
| | - Ragdha Alzarzour
- Discipline of Pharmacology, School of Pharmacy, Arab International University (AIU), Damascus, Syria
| | - Aidiahmad Dewa
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia
| | - Azira Muhamad
- Malaysia Genome and Vaccine Institute, National Institutes of Biotechnology Malaysia (NIBM), 43000 Bangi, Selangor, Malaysia
| | - Fauziahanim Zakaria
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia.
| |
Collapse
|
2
|
de Lange M, Yarosh V, Farell K, McDonnell C, Patil R, Hawthorn I, Jung MM, Wenje S, Steinert JR. High fat diet induces differential age- and gender-dependent changes in neuronal function in Drosophila linked to redox stress. Behav Brain Res 2025; 484:115510. [PMID: 40010512 DOI: 10.1016/j.bbr.2025.115510] [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: 10/30/2024] [Revised: 01/22/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025]
Abstract
The prevalence of neurodegenerative diseases, such as Alzheimer's and Parkinson's disease, is steadily increasing, thus posing significant challenges to global healthcare systems. Emerging evidence suggests that dietary habits, particularly consumption of high-fat diets, may play a pivotal role in the development and progression of neurodegenerative disorders. Moreover, several studies have shed light on the intricate communication between the gut and the brain, linking gut health with neuroinflammation and its involvement in neurodegenerative processes. This study aims to assess the effects of a high-fat dietary intake on various aspects of neuronal function during aging in a gender specific manner to help understand the potential contributions of diet to neuronal function. To investigate the effects of a high-fat diet, Drosophila melanogaster was used and exposed to a standard normal food diet (NF) and a high-fat diet (HF). Adults were grouped at 10 and 45 days of age in male and female flies reared under the same conditions starting the HF diet at 5 days of age with data showing differential gender- and HF diet-induced phenotypes. Malondialdehyde (MDA) levels were higher in males at 10 and 45 days (p < 0.05), caspase-3 expression increased at 45 days (p < 0.01) implicating apoptosis induction and a reduced climbing activity at 10 and 45 days was apparent in females only (p < 0.01). Adult lifespan under both dietary conditions was unchanged when reared at 18°C but odour-associated learning ability was reduced in larvae reared in a HF diet throughout their development (p < 0.05). This is the first study to characterise effects of a HF diet on neuronal phenotypes in an age- and gender-specific manner in a Drosophila model. Our findings suggest a HF diet induces differential effects of neuronal dysfunction with age and sex-specific outcomes, characterised by enhanced oxidative stress and cell death impacting on behaviour.
Collapse
Affiliation(s)
- Megan de Lange
- Division of Physiology, Pharmacology and Neuroscience, University of Nottingham, School of Life Sciences, Nottingham NG7 2NR, UK
| | - Vladyslava Yarosh
- Division of Physiology, Pharmacology and Neuroscience, University of Nottingham, School of Life Sciences, Nottingham NG7 2NR, UK
| | - Kevin Farell
- Division of Physiology, Pharmacology and Neuroscience, University of Nottingham, School of Life Sciences, Nottingham NG7 2NR, UK
| | - Caitlin McDonnell
- Division of Physiology, Pharmacology and Neuroscience, University of Nottingham, School of Life Sciences, Nottingham NG7 2NR, UK
| | - Renee Patil
- Division of Physiology, Pharmacology and Neuroscience, University of Nottingham, School of Life Sciences, Nottingham NG7 2NR, UK
| | - Isabel Hawthorn
- Division of Physiology, Pharmacology and Neuroscience, University of Nottingham, School of Life Sciences, Nottingham NG7 2NR, UK
| | - Mok-Min Jung
- Division of Physiology, Pharmacology and Neuroscience, University of Nottingham, School of Life Sciences, Nottingham NG7 2NR, UK
| | - Sophie Wenje
- Division of Physiology, Pharmacology and Neuroscience, University of Nottingham, School of Life Sciences, Nottingham NG7 2NR, UK
| | - Joern R Steinert
- Division of Physiology, Pharmacology and Neuroscience, University of Nottingham, School of Life Sciences, Nottingham NG7 2NR, UK.
| |
Collapse
|
3
|
Li HD, Zheng JY, Tan KW, Su JX, Chen W, Pang RK, Wu GL, Qiu YH, Li XX, Cai YF, Zhang SJ. Salvianolic acid B (SalB) improves high-fat diet (HFD)-caused cognitive impairment in mice by modulating the Trem2/Dap12 pathway in vivo and in vitro. Int Immunopharmacol 2025; 153:114461. [PMID: 40101423 DOI: 10.1016/j.intimp.2025.114461] [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: 12/13/2024] [Revised: 02/23/2025] [Accepted: 03/08/2025] [Indexed: 03/20/2025]
Abstract
Salvianolic acid B (SalB), which extracted from Salvia miltiorrhiza Bunge (Labiatae), is a traditional Chinese medicine. SalB is widely used in nervous system diseases. This study evaluated the protective effect of SalB on high-fat diet (HFD)-induced cognitive impairment and its mechanisms in vivo and in vitro. The behavior tests demonstrated that SalB alleviated motor skills and learning capacity in HFD mice. Animal experiments have confirmed that SalB reduced the mRNA expression of inflammatory markers and the Trem2/Dap12 pathway in HIP. Furthermore, SalB inhibited the microglia Trem2/Dap12 pathway in HIP. In vivo, palmitic acid (PA) was used to intervene in BV2 cells to construct an inflammatory. SalB reduced the mRNA expression of inflammatory markers and inhibited the Trem2/Dap12 pathway in BV2 cells. In conclusion, SalB treatment may serve as a possible therapy for cognitive impairment induced by HFD.
Collapse
Affiliation(s)
- Hong-Dan Li
- Department of Neurology, Nanning Hospital of Traditional Chinese Medicine, Nanning 530000, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Jia-Yi Zheng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Kai-Wen Tan
- Department of Neurology, Nanning Hospital of Traditional Chinese Medicine, Nanning 530000, China
| | - Jin-Xun Su
- Department of Neurology, Nanning Hospital of Traditional Chinese Medicine, Nanning 530000, China
| | - Wei Chen
- Department of Neurology, Nanning Hospital of Traditional Chinese Medicine, Nanning 530000, China
| | - Rui-Kang Pang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Guang-Liang Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Yu-Hui Qiu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Xiao-Xiao Li
- Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong 999077, China; State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China.
| | - Ye-Feng Cai
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China.
| | - Shi-Jie Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China.
| |
Collapse
|
4
|
Jost Z, Kujach S. Understanding Cognitive Decline in Aging: Mechanisms and Mitigation Strategies - A Narrative Review. Clin Interv Aging 2025; 20:459-469. [PMID: 40256418 PMCID: PMC12009036 DOI: 10.2147/cia.s510670] [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: 12/06/2024] [Accepted: 03/15/2025] [Indexed: 04/22/2025] Open
Abstract
Cognitive decline is a natural process that accompanies aging. In some cases, such as in sarcopenia-burdened or diseased older adults, the disease course may be more rapid. Declining cognitive function is associated with changes in the central nervous system per se or peripheral triggers that impair cognition. This review discusses issues related to central, central-peripheral, and peripheral factors that enhance cognitive deterioration, such as cortical thickness, cerebral white matter structure and function, blood-brain barrier (BBB) disruption, insulin resistance, inflammation, and vascular dysfunction. BBB permeability appears to be a critical point for factors associated with aging that may accelerate cognitive decline. Thus, we provide an in-depth analysis of the central-peripheral crosstalk. Additionally, we discuss high-intensity interval training (HIIT) as a promising strategy to counteract changes that accompany the aging process. Resistance (RHIIT) and aerobic (AHIIT) may be beneficial for cognitive health among the elderly, but their lack of empirical confirmation is a huge gap in the research.
Collapse
Affiliation(s)
- Zbigniew Jost
- Department of Biochemistry, Gdansk University of Physical Education and Sport, Gdansk, Poland
| | - Sylwester Kujach
- Department of Physiology, Medical University of Gdansk, Gdansk, Poland
- Department of Physiology, Gdansk University of Physical Education and Sport, Gdansk, Poland
| |
Collapse
|
5
|
Babakhani K, Kucinskas AL, Ye X, Giles ED, Sun Y. Aging immunity: unraveling the complex nexus of diet, gut microbiome, and immune function. IMMUNOMETABOLISM (COBHAM, SURREY) 2025; 7:e00061. [PMID: 40352822 PMCID: PMC12063687 DOI: 10.1097/in9.0000000000000061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/28/2025] [Indexed: 05/14/2025]
Abstract
Aging is associated with immune senescence and gut dysbiosis, both of which are heavily influenced by the diet. In this review, we summarize current knowledge regarding the impact of diets high in fiber, protein, or fat, as well as different dietary components (tryptophan, omega-3 fatty acids, and galacto-oligosaccharides) on the immune system and the gut microbiome in aging. Additionally, this review discusses how aging alters tryptophan metabolism, contributing to changes in immune function and the gut microbiome. Understanding the relationship between diet, the gut microbiome, and immune function in the context of aging is critical to formulate sound dietary recommendations for older individuals, and these personalized nutritional practices will ultimately improve the health and longevity of the elderly.
Collapse
Affiliation(s)
| | - Amanda L. Kucinskas
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Xiangcang Ye
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Erin D. Giles
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| |
Collapse
|
6
|
Song M, Bai Y, Song F. High-fat diet and neuroinflammation: The role of mitochondria. Pharmacol Res 2025; 212:107615. [PMID: 39842474 DOI: 10.1016/j.phrs.2025.107615] [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: 08/15/2024] [Revised: 12/28/2024] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
In recent years, increasing evidence has supported that high-fat diet (HFD) can induce the chronic, low-grade neuroinflammation in the brain, which is closely associated with the impairment of cognitive function. As the key organelles responsible for energy metabolism in the cell, mitochondria are believed to involved in the pathogenesis of a variety of neurological disorders. This review summarizes the current progress in the field of the relationship between HFD exposure and neurodegenerative diseases, and outline the major routines of HFD induced neuroinflammation and its pathological significance in the pathogenesis of neurodegenerative diseases. Furthermore, the article highlights the pivotal role of mitochondrial dysfunction in driving the neuroinflammation in the setting of HFD. Danger-associated molecular patterns (DAMPs) from damaged mitochondria can activate innate immune signaling pathways, while mitochondrial dysfunction itself can lead to metabolic remodeling of inflammatory cells, thus inducing neuroinflammation. More importantly, mitochondrial damage, neuroinflammation, and insulin resistance caused by HFD form a mutually reinforcing vicious cycle, ultimately leading to the death of neurons and promoting the progression of neurodegenerative diseases. Thus, in-depth elucidation of the role and underlying mechanisms of mitochondrial dysfunction in HFD-induced metabolic disorders may not only expand our understanding of the mechanistic linkages between HFD and etiology of neurodegenerative diseases, but also help develop the specific strategies for the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Mingxue Song
- Department of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China.
| | - Yao Bai
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100021, China.
| | - Fuyong Song
- Department of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China.
| |
Collapse
|
7
|
Jantsch J, da Silva Rodrigues F, Silva Dias V, de Farias Fraga G, Eller S, Giovenardi M, Guedes RP. Calorie Restriction Attenuates Memory Impairment and Reduces Neuroinflammation in Obese Aged Rats. Mol Neurobiol 2025; 62:1788-1799. [PMID: 39037530 DOI: 10.1007/s12035-024-04360-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024]
Abstract
Obesity and aging collectively potentiate inflammatory responses, particularly within the central nervous system. Managing obesity presents a significant challenge, even more so considering the context of aging. Caloric restriction (CR) has been extensively documented in the literature for its multiple health benefits. Motivated by these findings, we hypothesized that CR could serve as a valuable intervention to address the brain alterations and cognitive decline associated with obesity in aged rats. Our investigation revealed that cafeteria diet increased hippocampal and hypothalamic transcripts related to neuroinflammation, along with cognitive deficits determined in the object recognition test in 18-month-old male rats. Western blot data indicate that the obesogenic diet may disrupt the blood-brain barrier and lead to an increase in Toll-like receptor 4 in the hippocampus, events that could contribute to the cognitive deficits observed. Implementing CR after the onset of obesity mitigated neuroinflammatory changes and cognitive impairments. We found that CR increases GABA levels in the hippocampus of aged animals, as demonstrated by liquid chromatography coupled with mass spectrometry analysis. These findings underscore the potential of CR as a therapeutic opportunity to ameliorate the neuroinflammatory and cognitive alterations of obesity, especially in the context of aging.
Collapse
Affiliation(s)
- Jeferson Jantsch
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
| | - Fernanda da Silva Rodrigues
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
| | - Victor Silva Dias
- Biomedical Science Undergraduate Program, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
| | - Gabriel de Farias Fraga
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
| | - Sarah Eller
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
| | - Márcia Giovenardi
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
| | - Renata Padilha Guedes
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil.
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil.
| |
Collapse
|
8
|
Zeng J, Cheong LYT, Lo CH. Therapeutic targeting of obesity-induced neuroinflammation and neurodegeneration. Front Endocrinol (Lausanne) 2025; 15:1456948. [PMID: 39897964 PMCID: PMC11781992 DOI: 10.3389/fendo.2024.1456948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 12/30/2024] [Indexed: 02/04/2025] Open
Abstract
Obesity is a major modifiable risk factor leading to neuroinflammation and neurodegeneration. Excessive fat storage in obesity promotes the progressive infiltration of immune cells into adipose tissue, resulting in the release of pro-inflammatory factors such as cytokines and adipokines. These inflammatory mediators circulate through the bloodstream, propagating inflammation both in the periphery and in the central nervous system. Gut dysbiosis, which results in a leaky intestinal barrier, exacerbates inflammation and plays a significant role in linking obesity to the pathogenesis of neuroinflammation and neurodegeneration through the gut-brain/gut-brain-liver axis. Inflammatory states within the brain can lead to insulin resistance, mitochondrial dysfunction, autolysosomal dysfunction, and increased oxidative stress. These disruptions impair normal neuronal function and subsequently lead to cognitive decline and motor deficits, similar to the pathologies observed in major neurodegenerative diseases, including Alzheimer's disease, multiple sclerosis, and Parkinson's disease. Understanding the underlying disease mechanisms is crucial for developing therapeutic strategies to address defects in these inflammatory and metabolic pathways. In this review, we summarize and provide insights into different therapeutic strategies, including methods to alter gut dysbiosis, lifestyle changes, dietary supplementation, as well as pharmacological agents derived from natural sources, that target obesity-induced neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Jialiu Zeng
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, United States
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, United States
| | - Lenny Yi Tong Cheong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Chih Hung Lo
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, United States
- Department of Biology, Syracuse University, Syracuse, NY, United States
| |
Collapse
|
9
|
Butler MJ, Muscat SM, Caetano-Silva ME, Shrestha A, Olmo BMG, Mackey-Alfonso SE, Massa N, Alvarez BD, Blackwell JA, Bettes MN, DeMarsh JW, McCusker RH, Allen JM, Barrientos RM. Obesity-associated memory impairment and neuroinflammation precede widespread peripheral perturbations in aged rats. Immun Ageing 2025; 22:2. [PMID: 39754121 PMCID: PMC11697663 DOI: 10.1186/s12979-024-00496-3] [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: 10/25/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND Obesity and metabolic syndrome are major public health concerns linked to cognitive decline with aging. Prior work from our lab has demonstrated that short-term high fat diet (HFD) rapidly impairs memory function via a neuroinflammatory mechanism. However, the degree to which these rapid inflammatory changes are unique to the brain is unknown. Moreover, deviations in gut microbiome composition have been associated with obesity and cognitive impairment, but how diet and aging interact to impact the gut microbiome, or how rapidly these changes occur, is less clear. Thus, our study investigated the impact of HFD after two distinct consumption durations: 3 months (to model diet-induced obesity) or 3 days (to detect the rapid changes occurring with HFD) on memory function, anxiety-like behavior, central and peripheral inflammation, and gut microbiome profile in young and aged rats. RESULTS Our data indicated that both short-term and long-term HFD consumption impaired memory function and increased anxiety-like behavior in aged, but not young adult, rats. These behavioral changes were accompanied by pro- and anti-inflammatory cytokine dysregulation in the hippocampus and amygdala of aged HFD-fed rats at both time points. However, changes to fasting glucose, insulin, and inflammation in peripheral tissues such as the distal colon and visceral adipose tissue were increased in young and aged rats only after long-term, but not short-term, HFD consumption. Furthermore, while subtle HFD-induced changes to the gut microbiome did occur rapidly, robust age-specific effects were only present following long-term HFD consumption. CONCLUSIONS Overall, these data suggest that HFD-evoked neuroinflammation, memory impairment, and anxiety-like behavior in aging develop quicker than, and separately from the peripheral hallmarks of diet-induced obesity.
Collapse
Affiliation(s)
- Michael J Butler
- Institute for Behavioral Medicine Research, Ohio State University, 460 Medical Center Drive, Columbus, OH, 43210, USA
| | - Stephanie M Muscat
- Institute for Behavioral Medicine Research, Ohio State University, 460 Medical Center Drive, Columbus, OH, 43210, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | | | - Akriti Shrestha
- Department of Health and Kinesiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Brigitte M González Olmo
- Institute for Behavioral Medicine Research, Ohio State University, 460 Medical Center Drive, Columbus, OH, 43210, USA
| | - Sabrina E Mackey-Alfonso
- Institute for Behavioral Medicine Research, Ohio State University, 460 Medical Center Drive, Columbus, OH, 43210, USA
- Medical Scientist Training Program, The Ohio State University, Columbus, OH, USA
| | - Nashali Massa
- Institute for Behavioral Medicine Research, Ohio State University, 460 Medical Center Drive, Columbus, OH, 43210, USA
| | - Bryan D Alvarez
- Institute for Behavioral Medicine Research, Ohio State University, 460 Medical Center Drive, Columbus, OH, 43210, USA
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Jade A Blackwell
- Institute for Behavioral Medicine Research, Ohio State University, 460 Medical Center Drive, Columbus, OH, 43210, USA
- MCDB Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Menaz N Bettes
- Institute for Behavioral Medicine Research, Ohio State University, 460 Medical Center Drive, Columbus, OH, 43210, USA
| | - James W DeMarsh
- Institute for Behavioral Medicine Research, Ohio State University, 460 Medical Center Drive, Columbus, OH, 43210, USA
| | - Robert H McCusker
- Department of Health and Kinesiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jacob M Allen
- Department of Health and Kinesiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, Ohio State University, 460 Medical Center Drive, Columbus, OH, 43210, USA.
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA.
- Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, 460 Medical Center Drive, OH, 43210, USA.
- Chronic Brain Injury Program, Discovery Themes Initiative, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
10
|
Solch-Ottaiano RJ, Engler-Chiurazzi EB, Harper C, Wasson S, Ogbonna S, Ouvrier B, Wang H, Prats M, McDonald K, Biose IJ, Rowe LA, Jones M, Steele C, Bix G, Maraganore DM. Comparison Between Two Divergent Diets, Mediterranean and Western, on Gut Microbiota and Cognitive Function in Young Sprague Dawley Rats. GUT MICROBES REPORTS 2024; 1:1-21. [PMID: 39916748 PMCID: PMC11800364 DOI: 10.1080/29933935.2024.2439490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 11/19/2024] [Accepted: 12/02/2024] [Indexed: 02/09/2025]
Abstract
Clinical studies strongly suggest the importance of diet quality on cognition in youth populations (15-24 years). The Mediterranean diet (MeDi) has been shown to improve cognition in contrast to the commonly consumed Western diet (WD). The gut microbiota may serve as a mechanism for diet-induced changes in cognition. Ten-week-old male Sprague Dawley rats were assigned a MeDi or WD (n=10/group) for 14 weeks. Prior to neurobehavior assessments, microbiota community composition was assessed. At the genus level, the relative abundance of four bacteria increased with the MeDi and five decreased compared to the WD. Rats in the MeDi group demonstrated cognitive flexibility and improvement in reference and working memory relative to the WD group. At the end of the study, serum cytokines were increased, and low-density lipoproteins were decreased in the MeDi group. Markers for neuroinflammation, blood-brain barrier, glial cells, and synaptic plasticity in brain regions did not differ between groups. Overall, the MeDi modulated gut microbiota, cognitive function, and serum lipid and cytokines but not gene expression in the brain compared to the WD. Further studies are needed to determine causality between diet-modulated gut microbiota, cognitive function, and immune function.
Collapse
Affiliation(s)
- Rebecca J. Solch-Ottaiano
- Clinical Neuroscience Research Center, Department of Neurology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Elizabeth B. Engler-Chiurazzi
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Colin Harper
- Clinical Neuroscience Research Center, Department of Neurology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
| | - Savannah Wasson
- Clinical Neuroscience Research Center, Department of Neurology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Sharon Ogbonna
- Clinical Neuroscience Research Center, Department of Neurology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Blake Ouvrier
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Hanyun Wang
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Madison Prats
- Clinical Neuroscience Research Center, Department of Neurology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
| | - Katherine McDonald
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ifechukwude J. Biose
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Lori A. Rowe
- Virus Characterization, Isolation, Production and Sequencing Core, Department of Microbiology, Tulane National Primate Center, Covington, LA, USA
| | - MaryJane Jones
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Chad Steele
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Gregory Bix
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Demetrius M. Maraganore
- Clinical Neuroscience Research Center, Department of Neurology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
| |
Collapse
|
11
|
Drougard A, Ma EH, Wegert V, Sheldon R, Panzeri I, Vatsa N, Apostle S, Fagnocchi L, Schaf J, Gossens K, Völker J, Pang S, Bremser A, Dror E, Giacona F, Sagar S, Henderson MX, Prinz M, Jones RG, Pospisilik JA. An acute microglial metabolic response controls metabolism and improves memory. eLife 2024; 12:RP87120. [PMID: 39625057 PMCID: PMC11614388 DOI: 10.7554/elife.87120] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2024] Open
Abstract
Chronic high-fat feeding triggers metabolic dysfunction including obesity, insulin resistance, and diabetes. How high-fat intake first triggers these pathophysiological states remains unknown. Here, we identify an acute microglial metabolic response that rapidly translates intake of high-fat diet (HFD) to a surprisingly beneficial effect on metabolism and spatial/learning memory. High-fat intake rapidly increases palmitate levels in cerebrospinal fluid and triggers a wave of microglial metabolic activation characterized by mitochondrial membrane activation and fission as well as metabolic skewing toward aerobic glycolysis. These effects are detectable throughout the brain and can be detected within as little as 12 hr of HFD exposure. In vivo, microglial ablation and conditional DRP1 deletion show that the microglial metabolic response is necessary for the acute effects of HFD. 13C-tracing experiments reveal that in addition to processing via β-oxidation, microglia shunt a substantial fraction of palmitate toward anaplerosis and re-release of bioenergetic carbons into the extracellular milieu in the form of lactate, glutamate, succinate, and intriguingly, the neuroprotective metabolite itaconate. Together, these data identify microglia as a critical nutrient regulatory node in the brain, metabolizing away harmful fatty acids and liberating the same carbons as alternate bioenergetic and protective substrates for surrounding cells. The data identify a surprisingly beneficial effect of short-term HFD on learning and memory.
Collapse
Affiliation(s)
- Anne Drougard
- Department of Epigenetics, Van Andel Research InstituteGrand RapidsUnited States
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Eric H Ma
- Department of Metabolism and Nutritional Programming, Van Andel Research InstituteGrand RapidsUnited States
| | - Vanessa Wegert
- Department of Epigenetics, Van Andel Research InstituteGrand RapidsUnited States
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Ryan Sheldon
- Metabolomics and Bioenergetics Core, Van Andel InstituteGrand RapidsUnited States
| | - Ilaria Panzeri
- Department of Epigenetics, Van Andel Research InstituteGrand RapidsUnited States
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Naman Vatsa
- Department of Neurodegenerative Sciences, Van Andel Research InstituteGrand RapidsUnited States
| | - Stefanos Apostle
- Department of Epigenetics, Van Andel Research InstituteGrand RapidsUnited States
| | - Luca Fagnocchi
- Department of Epigenetics, Van Andel Research InstituteGrand RapidsUnited States
| | - Judith Schaf
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Klaus Gossens
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Josephine Völker
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Shengru Pang
- Institute of Neuropathology, Medical Faculty, University of FreiburgFreiburgGermany
| | - Anna Bremser
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Erez Dror
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Francesca Giacona
- Department of Epigenetics, Van Andel Research InstituteGrand RapidsUnited States
| | - Sagar Sagar
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
- Department of Medicine II, University Hospital FreiburgFreiburgGermany
| | - Michael X Henderson
- Department of Neurodegenerative Sciences, Van Andel Research InstituteGrand RapidsUnited States
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of FreiburgFreiburgGermany
- Centre for NeuroModulation (NeuroModBasics), University of FreiburgFreiburgGermany
- Signaling Research Centers BIOSS and CIBSS, University of FreiburgFreiburgGermany
| | - Russell G Jones
- Department of Metabolism and Nutritional Programming, Van Andel Research InstituteGrand RapidsUnited States
| | - John Andrew Pospisilik
- Department of Epigenetics, Van Andel Research InstituteGrand RapidsUnited States
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| |
Collapse
|
12
|
Oue K, Yamawaki Y, Ouhara K, Imado E, Tamura T, Doi M, Shimizu Y, Yoshida M, Mizuno N, Morioka N, Kanematsu T, Irifune M, Ago Y. Oral administration of Porphyromonas gingivalis to mice with diet-induced obesity impairs cognitive function associated with microglial activation in the brain. J Oral Microbiol 2024; 16:2419155. [PMID: 39553478 PMCID: PMC11565673 DOI: 10.1080/20002297.2024.2419155] [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: 03/27/2023] [Revised: 01/04/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024] Open
Abstract
Objective Both periodontal disease and obesity are risk factors for dementia, but their links to 1brain function remain unclear. In this study, we examined the effects of oral infection with a periodontal pathogen on cognitive function in a mouse model of obesity, focusing on the roles of microglia. Methods To create a mouse model of diet-induced obesity and periodontitis, male C57BL/6 J mice were first fed a high-fat diet containing 60% lipid calories for 18 weeks, beginning at 12 weeks of age, to achieve diet-induced obesity. Then, Porphyromonas gingivalis administration in the oral cavity twice weekly for 6 weeks was performed to induce periodontitis in obese mice. Results Obese mice orally exposed to P. gingivalis showed cognitive impairment in the novel object recognition test. Increased expression levels of inflammatory cytokines (e.g. interleukin-1β and tumor necrosis factor-α) were observed in the hippocampus of P. gingivalis-treated obese mice. Immunohistochemical analysis revealed that microglia cell body size was increased in the hippocampus and prefrontal cortex of P. gingivalis-treated obese mice, indicating microglial activation. Furthermore, depletion of microglia by PLX3397, a colony-stimulating factor 1 receptor inhibitor, ameliorated cognitive dysfunction. Conclusion These results suggest that microglia mediate periodontal infection-induced cognitive dysfunction in obesity.
Collapse
Affiliation(s)
- Kana Oue
- Department of Dental Anesthesiology, Division of Oral and Maxillofacial Surgery and Oral Medicine, Hiroshima University Hospital, Hiroshima, Japan
| | - Yosuke Yamawaki
- Department of Advanced Pharmacology, Daiichi University of Pharmacy, Fukuoka, Japan
| | - Kazuhisa Ouhara
- Department of Periodontal Medicine, Division of Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Eiji Imado
- Department of Dental Anesthesiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tetsuya Tamura
- Department of Periodontal Medicine, Division of Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mitsuru Doi
- Department of Dental Anesthesiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yoshitaka Shimizu
- Department of Dental Anesthesiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mitsuhiro Yoshida
- Department of Dental Anesthesiology, Division of Oral and Maxillofacial Surgery and Oral Medicine, Hiroshima University Hospital, Hiroshima, Japan
- Department of Dental Anesthesiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Noriyoshi Mizuno
- Department of Periodontal Medicine, Division of Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Norimitsu Morioka
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takashi Kanematsu
- Department of Cell Biology, Aging Science, and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Masahiro Irifune
- Department of Dental Anesthesiology, Division of Oral and Maxillofacial Surgery and Oral Medicine, Hiroshima University Hospital, Hiroshima, Japan
- Department of Dental Anesthesiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yukio Ago
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
13
|
Mackey-Alfonso SE, Butler MJ, Taylor AM, Williams-Medina AR, Muscat SM, Fu H, Barrientos RM. Short-term high fat diet impairs memory, exacerbates the neuroimmune response, and evokes synaptic degradation via a complement-dependent mechanism in a mouse model of Alzheimer's disease. Brain Behav Immun 2024; 121:56-69. [PMID: 39043341 DOI: 10.1016/j.bbi.2024.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 07/25/2024] Open
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disease characterized by profound memory impairments, synaptic loss, neuroinflammation, and hallmark pathological markers. High-fat diet (HFD) consumption increases the risk of developing AD even after controlling for metabolic syndrome, pointing to a role of the diet itself in increasing risk. In AD, the complement system, an arm of the immune system which normally tags redundant or damaged synapses for pruning, becomes pathologically overactivated leading to tagging of healthy synapses. While the unhealthy diet to AD link is strong, the underlying mechanisms are not well understood in part due to confounding variables associated with long-term HFD which can independently influence the brain. Therefore, we experimented with a short-term diet regimen to isolate the diet's impact on brain function without causing obesity. This project investigated the effect of short-term HFD on 1) memory, 2) neuroinflammation including complement, 3) AD pathology markers, 4) synaptic markers, and 5) in vitro microglial synaptic phagocytosis in the 3xTg-AD mouse model. Following the consumption of either standard chow or HFD, 3xTg-AD and non-Tg mice were tested for memory impairments. In a separate cohort of mice, levels of hippocampal inflammatory markers, complement proteins, AD pathology markers, and synaptic markers were measured. For the last set of experiments, BV2 microglial phagocytosis of synapses was evaluated. Synaptoneurosomes isolated from the hippocampus of 3xTg-AD mice fed chow or HFD were incubated with equal numbers of BV2 microglia. The number of BV2 microglia that phagocytosed synaptoneurosomes was tracked over time with a live-cell imaging assay. Finally, we incubated BV2 microglia with a complement receptor inhibitor (NIF) and repeated the assay. Behavioral analysis showed 3xTg-AD mice had significantly impaired long-term contextual and cued fear memory compared to non-Tg mice that was further impaired by HFD. HFD significantly increased inflammatory markers and complement expression while decreasing synaptic marker expression only in 3xTg-AD mice, without altering AD pathology markers. Synaptoneurosomes from HFD-fed 3xTg-AD mice were phagocytosed at a significantly higher rate than those from chow-fed mice, suggesting the synapses were altered by HFD. The complement receptor inhibitor blocked this effect in a dose-dependent manner, demonstrating the HFD-mediated increase in phagocytosis was complement dependent. This study indicates HFD consumption increases neuroinflammation and over-activates the complement cascade in 3xTg-AD mice, resulting in poorer memory. The in vitro data point to complement as a potential mechanistic culprit and therapeutic target underlying HFD's influence in increasing cognitive vulnerability to AD.
Collapse
Affiliation(s)
- Sabrina E Mackey-Alfonso
- Medical Scientist Training Program, The Ohio State University, Columbus, OH, USA; Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA; Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Michael J Butler
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA; Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, USA
| | - Ashton M Taylor
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | | | - Stephanie M Muscat
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Hongjun Fu
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA; Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
14
|
Xie QY, Shu X, Liu T, Chen LL, Luo LP. Ethanol Extract of Propolis Attenuates Liver Lipid Metabolism Disorder in High-Fat Diet-Fed SAMP8 Mice. Mol Nutr Food Res 2024; 68:e2400297. [PMID: 39165040 DOI: 10.1002/mnfr.202400297] [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: 04/22/2024] [Revised: 07/28/2024] [Indexed: 08/22/2024]
Abstract
SCOPE The prevalence of high-fat diet (HFD) consumption is increasing among middle-aged and older adults, which accelerates the aging process of this population and is more likely to induce lipid metabolism disorders. But the alleviation of ethanolic extract of propolis (EEP) on lipid metabolism disorders during aging remains unclear. METHODS AND RESULTS This study assesseed the impact of EEP intervention (200 mg kg-1 bw) on aging and lipid metabolism disorders in HFD-fed senescence accelerate mouse prone 8 (SAMP8) mice. Findings indicate that EEP ameliorates hair luster degradation and weight gain, reduces systemic inflammation and metabolism levels, enhances hepatic antioxidant enzyme activities, and improves the hepatic expression of senescence-associated secretory phenotype and aging-related genes in HFD-fed SAMP8 mice. Histological staining demonstrates that EEP improves hepatic lipid deposition and inflammatory cell infiltration. Transcriptomic and lipidomic analysis reveal that EEP promotes fatty acid β-oxidation by activating PPAR pathway, resulting in reduced hepatic lipid deposition, and attenuates bile acid (BA) accumulation by improving BA metabolism, which were ensured through qPCR validation of key genes and immunoblot validation of key proteins. CONCLUSIONS : EEP can regulate lipid metabolic dysregulation during aging accompanied by an HFD, potentially delaying the onset and progression of age-related diseases. This provides new approach for supporting healthy aging.
Collapse
Affiliation(s)
- Quan-Yuan Xie
- School of Life Sciences, Nanchang University, Nanchang, 330031, China
| | - Xin Shu
- School of Life Sciences, Nanchang University, Nanchang, 330031, China
| | - Tao Liu
- Key Laboratory of Geriatric Nutrition and Health of the Ministry of Education, Beijing Technology and Business University, Beijing, 100048, China
| | - Li-Li Chen
- School of Life Sciences, Nanchang University, Nanchang, 330031, China
| | - Li-Ping Luo
- School of Life Sciences, Nanchang University, Nanchang, 330031, China
- Key Laboratory of Geriatric Nutrition and Health of the Ministry of Education, Beijing Technology and Business University, Beijing, 100048, China
| |
Collapse
|
15
|
Sanz-Martos AB, Roca M, Ruiz-Gayo M, Del Olmo N. Tributyrin reverses the deleterious effect of saturated fat on working memory and synaptic plasticity in juvenile mice: differential effects in males and females. Eur J Pharmacol 2024; 977:176726. [PMID: 38852700 DOI: 10.1016/j.ejphar.2024.176726] [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: 02/19/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/11/2024]
Abstract
Short-chain fatty acids, such as butyric acid, derived from the intestinal fermentation of dietary fiber, have been proposed as a treatment for certain pathologies of the central nervous system. Our research group has shown that tributyrin (TB), a butyric acid prodrug, reverses deficits in spatial memory and modulates hippocampal synaptic plasticity. In the present work, diets enriched in either saturated (SOLF; Saturated OiL-enriched Food) or unsaturated (UOLF; Unsaturated OiL-enriched Food) fat were supplied during either 2 h or 8 weeks to 5-week-old male and female mice undergoing a treatment schedule with TB. After the dietary treatment, spatial learning and memory (SLM) was assessed in both the Y-maze and the eight-arm radial maze (RAM). Hippocampal expression of genes involved in glutamatergic transmission as well as synaptic plasticity (long-term potentiation -LTP- and long-term depression -LTD-) were also analyzed. Our results show that 2 h of SOLF intake impaired LTP as well as the performance in the Y-Maze in juvenile male mice whereas no effect was found in females. Moreover, TB reversed both effects in SLM and LTP in males. In the case of chronic intake, both SOLF and UOLF deteriorated SLM measured in the RAM in both sexes whereas TB only reversed LTP impairment induced by SOLF in male mice. These results suggest that TB may have a potentially beneficial influence on learning and memory processes, contingent upon the type of diet and the sex of the individuals.
Collapse
Affiliation(s)
- Ana Belén Sanz-Martos
- Department of Psychobiology, School of Psychology, UNED, C/ Juan del Rosal 10, 28040, Madrid, Spain.
| | - María Roca
- Department of Psychobiology, School of Psychology, UNED, C/ Juan del Rosal 10, 28040, Madrid, Spain
| | - Mariano Ruiz-Gayo
- Department of Health and Pharmaceutical Sciences, School of Pharmacy, Universidad CEU-San Pablo, CEU Universities, 28668, Madrid, Spain
| | - Nuria Del Olmo
- Department of Psychobiology, School of Psychology, UNED, C/ Juan del Rosal 10, 28040, Madrid, Spain
| |
Collapse
|
16
|
Malik S, Xavier S, Soch A, Younesi S, Yip J, Slayo M, Barrientos RM, Sominsky L, Spencer SJ. High-fat diet and aging-associated memory impairments persist in the absence of microglia in female rats. Neurobiol Aging 2024; 140:22-32. [PMID: 38703636 DOI: 10.1016/j.neurobiolaging.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024]
Abstract
Aging is associated with a priming of microglia such that they are hypersensitive to further immune challenges. As such high-fat diet during aging can have detrimental effects on cognition that is not seen in the young. However, conflicting findings also suggest that obesity may protect against cognitive decline during aging. Given this uncertainty we aimed here to examine the role of microglia in high-fat, high-sucrose diet (HFSD)-induced changes in cognitive performance in the aging brain. We hypothesised that 8 weeks of HFSD-feeding would alter microglia and the inflammatory milieu in aging and worsen aging-related cognitive deficits in a microglia-dependent manner. We found that both aging and HFSD reduced hippocampal neuron numbers and open field exploration; they also impaired recognition memory. However, the aging-related deficits occurred in the absence of a pro-inflammatory response and the deficits in memory performance persisted after depletion of microglia in the Cx3cr1-Dtr knock-in rat. Our data suggest that mechanisms additional to the acute microglial contribution play a role in aging- and HFSD-associated memory dysfunction.
Collapse
Affiliation(s)
- Sajida Malik
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, Australia; IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Soniya Xavier
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, Australia
| | - Alita Soch
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, Australia; The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Simin Younesi
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, Australia; Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Jackson Yip
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, Australia
| | - Mary Slayo
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, Australia; Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Germany; Center for Mind, Brain and Behavior-CMBB, Giessen, Marburg, Germany
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA
| | - Luba Sominsky
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, Australia; Barwon Health, Geelong, Victoria, Australia; IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, Australia.
| |
Collapse
|
17
|
Zhang JS, Li S, Cheng X, Tan XC, Huang YL, Dong HJ, Xue R, Zhang Y, Li JC, Feng XX, Deng Y, Zhang YZ. Far-Infrared Therapy Based on Graphene Ameliorates High-Fat Diet-Induced Anxiety-Like Behavior in Obese Mice via Alleviating Intestinal Barrier Damage and Neuroinflammation. Neurochem Res 2024; 49:1735-1750. [PMID: 38530508 DOI: 10.1007/s11064-024-04133-9] [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: 11/26/2023] [Revised: 01/29/2024] [Accepted: 02/20/2024] [Indexed: 03/28/2024]
Abstract
The consumption of a high-fat diet (HFD) has been implicated in the etiology of obesity and various neuropsychiatric disturbances, including anxiety and depression. Compelling evidence suggests that far-infrared ray (FIR) possesses beneficial effects on emotional disorders. However, the efficacy of FIR therapy in addressing HFD-induced anxiety and the underlying mechanisms remain to be elucidated. Here, we postulate that FIR emitted from a graphene-based therapeutic device may mitigate HFD-induced anxiety behaviors. The graphene-FIR modify the gut microbiota in HFD-mice, particularly by an enriched abundance of beneficial bacteria Clostridiaceae and Erysipelotrichaceae, coupled with a diminution of harmful bacteria Lachnospiraceae, Anaerovoracaceae, Holdemania and Marvinbryantia. Graphene-FIR also improved intestinal barrier function, as evidenced by the augmented expression of the tight junction protein occludin and G protein-coupled receptor 43 (GPR43). In serum level, we observed the decreased free fatty acids (FFA), lipopolysaccharides (LPS), diamine oxidase (DAO) and D-lactate, and increased the glucagon-like peptide-2 (GLP-2) levels in graphene-FIR mice. Simultaneously, inflammatory cytokines IL-6, IL-1β, and TNF-α manifested a decrease subsequent to graphene-FIR treatment in both peripheral and central system. Notably, graphene-FIR inhibited over expression of astrocytes and microglia. We further noticed that the elevated the BDNF and decreased TLR4 and NF-κB expression in graphene-FIR group. Overall, our study reveals that graphene-FIR rescued HFD-induced anxiety via improving the intestine permeability and the integrity of blood-brain barrier, and reduced inflammatory response by down regulating TLR4/NF-κB inflammatory pathway.
Collapse
Affiliation(s)
- Jin-Shui Zhang
- School of Medicine, Anhui University of Science and Technology, Huainan, 232001, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Shuo Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Xin Cheng
- Hebei North University, Hebei, 075000, China
| | - Xiao-Cui Tan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Yu-Long Huang
- School of Medicine, Anhui University of Science and Technology, Huainan, 232001, China
| | - Hua-Jin Dong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Rui Xue
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Yang Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Jing-Cao Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Xiao-Xing Feng
- Grahope New Materials Technologies Inc., Shenzhen, 518063, China
| | - Yun Deng
- School of Medicine, Anhui University of Science and Technology, Huainan, 232001, China.
| | - You-Zhi Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| |
Collapse
|
18
|
N'Diaye M, Ducourneau EG, Bakoyiannis I, Potier M, Lafenetre P, Ferreira G. Obesogenic diet induces sex-specific alterations of contextual fear memory and associated hippocampal activity in mice. Cereb Cortex 2024; 34:bhae254. [PMID: 38934712 DOI: 10.1093/cercor/bhae254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
In addition to metabolic and cardiovascular disorders, obesity is associated with cognitive deficits in humans and animal models. We have previously shown that obesogenic high-fat and sugar diet intake during adolescence (adoHFSD) impairs hippocampus (HPC)-dependent memory in rodents. These results were obtained in males only and it remains to evaluate whether adoHFSD has similar effect in females. Therefore, here, we investigated the effects of adoHFSD consumption on HPC-dependent contextual fear memory and associated brain activation in male and female mice. Exposure to adoHFSD increased fat mass accumulation and glucose levels in both males and females but impaired contextual fear memory only in males. Compared with females, contextual fear conditioning induced higher neuronal activation in the dorsal and ventral HPC (CA1 and CA3 subfields) as well as in the medial prefrontal cortex in males. Also, adoHFSD-fed males showed enhanced c-Fos expression in the dorsal HPC, particularly in the dentate gyrus, and in the basolateral amygdala compared with the other groups. Finally, chemogenetic inactivation of the dorsal HPC rescued adoHFSD-induced memory deficits in males. Our results suggest that males are more vulnerable to the effects of adoHFSD on HPC-dependent aversive memory than females, due to overactivation of the dorsal HPC.
Collapse
Affiliation(s)
- Matéo N'Diaye
- NutriNeuro Lab, FoodCircus Team, Université de Bordeaux, UMR 1286 INRAE, Bordeaux INP, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Eva-Gunnel Ducourneau
- NutriNeuro Lab, FoodCircus Team, Université de Bordeaux, UMR 1286 INRAE, Bordeaux INP, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Ioannis Bakoyiannis
- NutriNeuro Lab, FoodCircus Team, Université de Bordeaux, UMR 1286 INRAE, Bordeaux INP, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Mylène Potier
- NutriNeuro Lab, FoodCircus Team, Université de Bordeaux, UMR 1286 INRAE, Bordeaux INP, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Pauline Lafenetre
- NutriNeuro Lab, FoodCircus Team, Université de Bordeaux, UMR 1286 INRAE, Bordeaux INP, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Guillaume Ferreira
- NutriNeuro Lab, FoodCircus Team, Université de Bordeaux, UMR 1286 INRAE, Bordeaux INP, 146 rue Léo Saignat, 33076 Bordeaux, France
| |
Collapse
|
19
|
Jonischkies K, del Angel M, Demiray YE, Loaiza Zambrano A, Stork O. The NDR family of kinases: essential regulators of aging. Front Mol Neurosci 2024; 17:1371086. [PMID: 38803357 PMCID: PMC11129689 DOI: 10.3389/fnmol.2024.1371086] [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: 01/15/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
Aging is defined as a progressive decline of cognitive and physiological functions over lifetime. Since the definition of the nine hallmarks of aging in 2013 by López-Otin, numerous studies have attempted to identify the main regulators and contributors in the aging process. One interesting group of proteins whose participation has been implicated in several aging hallmarks are the nuclear DBF2-related (NDR) family of serine-threonine AGC kinases. They are one of the core components of the Hippo signaling pathway and include NDR1, NDR2, LATS1 and LATS2 in mammals, along with its highly conserved metazoan orthologs; Trc in Drosophila melanogaster, SAX-1 in Caenorhabditis elegans, CBK1, DBF20 in Saccharomyces cerevisiae and orb6 in Saccharomyces pombe. These kinases have been independently linked to the regulation of widely diverse cellular processes disrupted during aging such as the cell cycle progression, transcription, intercellular communication, nutrient homeostasis, autophagy, apoptosis, and stem cell differentiation. However, a comprehensive overview of the state-of-the-art knowledge regarding the post-translational modifications of and by NDR kinases in aging has not been conducted. In this review, we summarize the current understanding of the NDR family of kinases, focusing on their relevance to various aging hallmarks, and emphasize the growing body of evidence that suggests NDR kinases are essential regulators of aging across species.
Collapse
Affiliation(s)
- Kevin Jonischkies
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Miguel del Angel
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Yunus Emre Demiray
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Allison Loaiza Zambrano
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Oliver Stork
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Science, Magdeburg, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
- German Center for Mental Health (DZPG), Jena-Magdeburg-Halle, Germany
| |
Collapse
|
20
|
Song M, Qiang Y, Wang S, Shan S, Zhang L, Liu C, Song F, Zhao X. High-fat diet exacerbates 1-Bromopropane-induced loss of dopaminergic neurons in the substantia nigra of mice through mitochondrial damage associated necroptotic pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 276:116280. [PMID: 38574648 DOI: 10.1016/j.ecoenv.2024.116280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 03/25/2024] [Accepted: 03/30/2024] [Indexed: 04/06/2024]
Abstract
In recent years, accumulating evidence supports that occupational exposure to solvents is associated with an increased incidence of Parkinson's disease (PD) among workers. The neurotoxic effects of 1-bromopropane (1-BP), a widely used new-type solvent, are well-established, yet data on its relationship with the etiology of PD remain limited. Simultaneously, high-fat consumption in modern society is recognized as a significant risk factor for PD. However, whether there is a synergistic effect between a high-fat diet and 1-BP exposure remains unclear. In this study, adult C57BL/6 mice were fed either a chow or a high-fat diet for 18 weeks prior to 12-week 1-BP treatment. Subsequent neurobehavioral and neuropathological examinations were conducted to assess the effects of 1-BP exposure on parkinsonian pathology. The results demonstrated that 1-BP exposure produced obvious neurobehavioral abnormalities and dopaminergic degeneration in the nigral region of mice. Importantly, a high-fat diet further exacerbated the impact of 1-BP on motor and cognitive abnormalities in mice. Mechanistic investigation revealed that mitochondrial damage and mtDNA release induced by 1-BP and high-fat diet activate NLRP3 and cGAS-STING pathway- mediated neuroinflammatory response, and ultimately lead to necroptosis of dopaminergic neurons. In summary, our study unveils a potential link between chronic 1-BP exposure and PD-like pathology with motor and no-motor defects in experimental animals, and long-term high-fat diet can further promote 1-BP neurotoxicity, which underscores the pivotal role of environmental factors in the etiology of PD.
Collapse
Affiliation(s)
- Mingxue Song
- Department of Health Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yalong Qiang
- Department of Health Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Shuai Wang
- Department of Health Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Shan Shan
- Department of Health Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Liwen Zhang
- Department of Health Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Caipei Liu
- Department of Health Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Fuyong Song
- Department of Health Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiulan Zhao
- Department of Health Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
21
|
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
|
22
|
Drougard A, Ma EH, Wegert V, Sheldon R, Panzeri I, Vatsa N, Apostle S, Fagnocchi L, Schaf J, Gossens K, Völker J, Pang S, Bremser A, Dror E, Giacona F, Sagar, Henderson MX, Prinz M, Jones RG, Pospisilik JA. An acute microglial metabolic response controls metabolism and improves memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.03.535373. [PMID: 37066282 PMCID: PMC10103996 DOI: 10.1101/2023.04.03.535373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Chronic high-fat feeding triggers chronic metabolic dysfunction including obesity, insulin resistance, and diabetes. How high-fat intake first triggers these pathophysiological states remains unknown. Here, we identify an acute microglial metabolic response that rapidly translates intake of high-fat diet (HFD) to a surprisingly beneficial effect on metabolism and spatial / learning memory. High-fat intake rapidly increases palmitate levels in cerebrospinal fluid and triggers a wave of microglial metabolic activation characterized by mitochondrial membrane activation and fission as well as metabolic skewing towards aerobic glycolysis. These effects are detectable throughout the brain and can be detected within as little as 12 hours of HFD exposure. In vivo, microglial ablation and conditional DRP1 deletion show that the microglial metabolic response is necessary for the acute effects of HFD. 13C-tracing experiments reveal that in addition to processing via β-oxidation, microglia shunt a substantial fraction of palmitate towards anaplerosis and re-release of bioenergetic carbons into the extracellular milieu in the form of lactate, glutamate, succinate, and intriguingly, the neuro-protective metabolite itaconate. Together, these data identify microglia as a critical nutrient regulatory node in the brain, metabolizing away harmful fatty acids and releasing the same carbons as alternate bioenergetic and protective substrates for surrounding cells. The data identify a surprisingly beneficial effect of short-term HFD on learning and memory.
Collapse
Affiliation(s)
- Anne Drougard
- Department of Epigenetics, Van Andel Research Institute, 333 Bostwick Ave, 49503, Grand Rapids, MI, USA
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Eric H Ma
- Department of Metabolism and Nutritional Programming, Van Andel Research Institute, 333 Bostwick Ave, 49503, Grand Rapids, MI, USA
| | - Vanessa Wegert
- Department of Epigenetics, Van Andel Research Institute, 333 Bostwick Ave, 49503, Grand Rapids, MI, USA
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Ryan Sheldon
- Metabolomics and Bioenergetics Core, Van Andel Research Institute, 333 Bostwick Ave, 49503, Grand Rapids, MI, USA
| | - Ilaria Panzeri
- Department of Epigenetics, Van Andel Research Institute, 333 Bostwick Ave, 49503, Grand Rapids, MI, USA
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Naman Vatsa
- Department of Neurodegenerative Sciences, Van Andel Research Institute, 333 Bostwick Ave, 49503, Grand Rapids, MI, USA
| | - Stefanos Apostle
- Department of Epigenetics, Van Andel Research Institute, 333 Bostwick Ave, 49503, Grand Rapids, MI, USA
| | - Luca Fagnocchi
- Department of Epigenetics, Van Andel Research Institute, 333 Bostwick Ave, 49503, Grand Rapids, MI, USA
| | - Judith Schaf
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Klaus Gossens
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Josephine Völker
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Shengru Pang
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Anna Bremser
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Erez Dror
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Francesca Giacona
- Department of Epigenetics, Van Andel Research Institute, 333 Bostwick Ave, 49503, Grand Rapids, MI, USA
| | - Sagar
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
- Department of Medicine II, University Hospital Freiburg, Freiburg, Germany
| | - Michael X Henderson
- Department of Neurodegenerative Sciences, Van Andel Research Institute, 333 Bostwick Ave, 49503, Grand Rapids, MI, USA
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Centre for NeuroModulation (NeuroModBasics), University of Freiburg, Freiburg, Germany
- Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Russell G Jones
- Department of Metabolism and Nutritional Programming, Van Andel Research Institute, 333 Bostwick Ave, 49503, Grand Rapids, MI, USA
| | - J. Andrew Pospisilik
- Department of Epigenetics, Van Andel Research Institute, 333 Bostwick Ave, 49503, Grand Rapids, MI, USA
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| |
Collapse
|
23
|
Kawade N, Yamanaka K. Novel insights into brain lipid metabolism in Alzheimer's disease: Oligodendrocytes and white matter abnormalities. FEBS Open Bio 2024; 14:194-216. [PMID: 37330425 PMCID: PMC10839347 DOI: 10.1002/2211-5463.13661] [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: 05/10/2023] [Revised: 06/07/2023] [Accepted: 06/14/2023] [Indexed: 06/19/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. A genome-wide association study has shown that several AD risk genes are involved in lipid metabolism. Additionally, epidemiological studies have indicated that the levels of several lipid species are altered in the AD brain. Therefore, lipid metabolism is likely changed in the AD brain, and these alterations might be associated with an exacerbation of AD pathology. Oligodendrocytes are glial cells that produce the myelin sheath, which is a lipid-rich insulator. Dysfunctions of the myelin sheath have been linked to white matter abnormalities observed in the AD brain. Here, we review the lipid composition and metabolism in the brain and myelin and the association between lipidic alterations and AD pathology. We also present the abnormalities in oligodendrocyte lineage cells and white matter observed in AD. Additionally, we discuss metabolic disorders, including obesity, as AD risk factors and the effects of obesity and dietary intake of lipids on the brain.
Collapse
Affiliation(s)
- Noe Kawade
- Department of Neuroscience and Pathobiology, Research Institute of Environmental MedicineNagoya UniversityJapan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of MedicineNagoya UniversityJapan
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental MedicineNagoya UniversityJapan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of MedicineNagoya UniversityJapan
- Institute for Glyco‐core Research (iGCORE)Nagoya UniversityJapan
- Center for One Medicine Innovative Translational Research (COMIT)Nagoya UniversityJapan
| |
Collapse
|
24
|
Lu T, Ding L, Zheng X, Li Y, Wei W, Liu W, Tao J, Xue X. Alisol A Exerts Neuroprotective Effects Against HFD-Induced Pathological Brain Aging via the SIRT3-NF-κB/MAPK Pathway. Mol Neurobiol 2024; 61:753-771. [PMID: 37659035 PMCID: PMC10861652 DOI: 10.1007/s12035-023-03592-5] [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: 01/17/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023]
Abstract
Chronic consumption of a high-fat diet (HFD) has profound effects on brain aging, which is mainly characterized by cognitive decline, inflammatory responses, and neurovascular damage. Alisol A (AA) is a triterpenoid with therapeutic potential for metabolic diseases, but whether it has a neuroprotective effect against brain aging caused by a HFD has not been investigated. Six-month-old male C57BL6/J mice were exposed to a HFD with or without AA treatment for 12 weeks. Behavioral tasks were used to assess the cognitive abilities of the mice. Neuroinflammation and changes in neurovascular structure in the brains were examined. We further assessed the mechanism by which AA exerts neuroprotective effects against HFD-induced pathological brain aging in vitro and in vivo. Behavioral tests showed that cognitive function was improved in AA-treated animals. AA treatment reduced microglia activation and inflammatory cytokine release induced by a HFD. Furthermore, AA treatment increased the number of hippocampal neurons, the density of dendritic spines, and the expression of tight junction proteins. We also demonstrated that AA attenuated microglial activation by targeting the SIRT3-NF-κB/MAPK pathway and ameliorated microglial activation-induced tight junction degeneration in endothelial cells and apoptosis in hippocampal neurons. The results of this study show that AA may be a promising agent for the treatment of HFD-induced brain aging.
Collapse
Affiliation(s)
- Taotao Lu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350112, China
- Fujian Key Laboratory of Rehabilitation Techniques, Cognitive Rehabilitation, Fuzhou, 350112, China
| | - Linlin Ding
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350112, China
| | - Xiaoqing Zheng
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350112, China
| | - Yongxu Li
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350112, China
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, No 13, Hudongzhi Road, Fuzhou City, 350003, Fujian Province, China
| | - Wei Wei
- Fujian Key Laboratory of Rehabilitation Techniques, Cognitive Rehabilitation, Fuzhou, 350112, China
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, No 13, Hudongzhi Road, Fuzhou City, 350003, Fujian Province, China
| | - Weilin Liu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350112, China
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350112, China
- Fujian Key Laboratory of Rehabilitation Techniques, Cognitive Rehabilitation, Fuzhou, 350112, China
| | - Xiehua Xue
- Fujian Key Laboratory of Rehabilitation Techniques, Cognitive Rehabilitation, Fuzhou, 350112, China.
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, No 13, Hudongzhi Road, Fuzhou City, 350003, Fujian Province, China.
| |
Collapse
|
25
|
Leuner B. Microglia and age-related cognitive decline: Primed for sex differences. Brain Behav Immun 2024; 116:267-268. [PMID: 38096924 DOI: 10.1016/j.bbi.2023.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 12/04/2023] [Indexed: 12/25/2023] Open
Affiliation(s)
- Benedetta Leuner
- Departments of Psychology and Neuroscience, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
26
|
Muscat SM, Butler MJ, Bettes MN, DeMarsh JW, Scaria EA, Deems NP, Barrientos RM. Post-operative cognitive dysfunction is exacerbated by high-fat diet via TLR4 and prevented by dietary DHA supplementation. Brain Behav Immun 2024; 116:385-401. [PMID: 38145855 PMCID: PMC10872288 DOI: 10.1016/j.bbi.2023.12.028] [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: 10/22/2023] [Revised: 12/04/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023] Open
Abstract
Post-operative cognitive dysfunction (POCD) is an abrupt decline in neurocognitive function arising shortly after surgery and persisting for weeks to months, increasing the risk of dementia diagnosis. Advanced age, obesity, and comorbidities linked to high-fat diet (HFD) consumption such as diabetes and hypertension have been identified as risk factors for POCD, although underlying mechanisms remain unclear. We have previously shown that surgery alone, or 3-days of HFD can each evoke sufficient neuroinflammation to cause memory deficits in aged, but not young rats. The aim of the present study was to determine if HFD consumption before surgery would potentiate and prolong the subsequent neuroinflammatory response and memory deficits, and if so, to determine the extent to which these effects depend on activation of the innate immune receptor TLR4, which both insults are known to stimulate. Young-adult (3mo) & aged (24mo) male F344xBN F1 rats were fed standard chow or HFD for 3-days immediately before sham surgery or laparotomy. In aged rats, the combination of HFD and surgery caused persistent deficits in contextual memory and cued-fear memory, though it was determined that HFD alone was sufficient to cause the long-lasting cued-fear memory deficits. In young adult rats, HFD + surgery caused only cued-fear memory deficits. Elevated proinflammatory gene expression in the hippocampus of both young and aged rats that received HFD + surgery persisted for at least 3-weeks after surgery. In a separate experiment, rats were administered the TLR4-specific antagonist, LPS-RS, immediately before HFD onset, which ameliorated the HFD + surgery-associated neuroinflammation and memory deficits. Similarly, dietary DHA supplementation for 4 weeks prior to HFD onset blunted the neuroinflammatory response to surgery and prevented development of persistent memory deficits. These results suggest that HFD 1) increases risk of persistent POCD-associated memory impairments following surgery in male rats in 2) a TLR4-dependent manner, which 3) can be targeted by DHA supplementation to mitigate development of persistent POCD.
Collapse
Affiliation(s)
- Stephanie M Muscat
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Michael J Butler
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Menaz N Bettes
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - James W DeMarsh
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Emmanuel A Scaria
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Nicholas P Deems
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Department of Psychiatry & Behavioral Health, The Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
27
|
Kolanu ND, Ahmed S, Kerimkulova MK, Stańczak M, Aguirre Vera GDJ, Shaikh N, Addula AR, Cheran M, Chilla SP, Oliveira Souza Lima SR, Shehryar A, Rehman A. Influence of Nutritional Interventions on Functional Outcomes in Stroke Rehabilitation: A Systematic Review. Cureus 2024; 16:e53711. [PMID: 38455777 PMCID: PMC10918289 DOI: 10.7759/cureus.53711] [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] [Accepted: 02/06/2024] [Indexed: 03/09/2024] Open
Abstract
Stroke, a major cause of disability worldwide, necessitates comprehensive rehabilitation, with nutrition playing a pivotal role in recovery. Our systematic review assesses the impact of nutritional interventions on stroke survivors' rehabilitation. Adhering to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we systematically searched PubMed, Embase, Web of Science, and Scopus, using keywords related to stroke rehabilitation and nutrition. Studies were selected based on criteria emphasizing dietary interventions and their effect on functional recovery in stroke patients. The review involved detailed data extraction and synthesis, covering study design, participant characteristics, interventions, and outcomes. Five studies met our inclusion criteria, encompassing longitudinal and prospective studies, retrospective cohorts, and randomized controlled trials. These studies highlighted the importance of early nutritional assessment, particularly for sarcopenic patients, and the role of energy and protein intake soon after a stroke. Findings indicated high nutritional risk correlated with poorer functional outcomes and increased inflammation. Tailored dietary support appeared beneficial for muscle mass maintenance and overall functional recovery, especially in older patients. Our review emphasizes the critical role of nutritional interventions in stroke rehabilitation. It suggests that personalized nutritional strategies can positively impact functional recovery, notably in older and nutritionally vulnerable stroke survivors. These insights underscore the necessity of integrating dietary assessments and interventions into standard stroke rehabilitation protocols, advocating a holistic approach to patient care.
Collapse
Affiliation(s)
| | - Sheraz Ahmed
- Medicine, Islamic International Medical College, Islamabad, PAK
| | | | - Mikołaj Stańczak
- Medicine, Kharkiv Institute of Medicine and Biomedical Sciences, Kharkiv, UKR
| | | | | | | | - Meher Cheran
- Internal Medicine, American International Medical University, Chicago, USA
| | - Srikar P Chilla
- Medicine, CARE Hospitals, Hyderabad, IND
- School of Health Sciences, University of East London, London, GBR
| | | | | | | |
Collapse
|
28
|
Vega-Rivera NM, Estrada-Camarena E, Azpilcueta-Morales G, Cervantes-Anaya N, Treviño S, Becerril-Villanueva E, López-Rubalcava C. Chronic Variable Stress and Cafeteria Diet Combination Exacerbate Microglia and c-fos Activation but Not Experimental Anxiety or Depression in a Menopause Model. Int J Mol Sci 2024; 25:1455. [PMID: 38338735 PMCID: PMC10855226 DOI: 10.3390/ijms25031455] [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: 12/06/2023] [Revised: 01/11/2024] [Accepted: 01/13/2024] [Indexed: 02/12/2024] Open
Abstract
The menopause transition is a vulnerable period for developing both psychiatric and metabolic disorders, and both can be enhanced by stressful events worsening their effects. The present study aimed to evaluate whether a cafeteria diet (CAF) combined with chronic variable stress (CVS) exacerbates anxious- or depressive-like behavior and neuronal activation, cell proliferation and survival, and microglia activation in middle-aged ovariectomized (OVX) rats. In addition, body weight, lipid profile, insulin resistance, and corticosterone as an index of metabolic changes or hypothalamus-pituitary-adrenal (HPA) axis activation, and the serum pro-inflammatory cytokines IL-6, IL-β, and TNFα were measured. A CAF diet increased body weight, lipid profile, and insulin resistance. CVS increased corticosterone and reduced HDL. A CAF produced anxiety-like behaviors, whereas CVS induced depressive-like behaviors. CVS increased serum TNFα independently of diet. A CAF and CVS separately enhanced the percentage of Iba-positive cells in the hippocampus; the combination of factors further increased Iba-positive cells in the ventral hippocampus. A CAF and CVS increased the c-fos-positive cells in the hippocampus; the combination of factors increased the number of positive cells expressing c-fos in the ventral hippocampus even more. The combination of a CAF and CVS generates a slight neuroinflammation process and neuronal activation in a hippocampal region-specific manner and differentially affects the behavior.
Collapse
Affiliation(s)
- Nelly Maritza Vega-Rivera
- Laboratorio de Neuropsicofarmacología, Dirección de Neurociencias, Instituto Nacional de Psiquiatría “Ramón de la Fuente”, Mexico City 14370, Mexico; (N.M.V.-R.); (G.A.-M.); (N.C.-A.)
| | - Erika Estrada-Camarena
- Laboratorio de Neuropsicofarmacología, Dirección de Neurociencias, Instituto Nacional de Psiquiatría “Ramón de la Fuente”, Mexico City 14370, Mexico; (N.M.V.-R.); (G.A.-M.); (N.C.-A.)
| | - Gabriel Azpilcueta-Morales
- Laboratorio de Neuropsicofarmacología, Dirección de Neurociencias, Instituto Nacional de Psiquiatría “Ramón de la Fuente”, Mexico City 14370, Mexico; (N.M.V.-R.); (G.A.-M.); (N.C.-A.)
| | - Nancy Cervantes-Anaya
- Laboratorio de Neuropsicofarmacología, Dirección de Neurociencias, Instituto Nacional de Psiquiatría “Ramón de la Fuente”, Mexico City 14370, Mexico; (N.M.V.-R.); (G.A.-M.); (N.C.-A.)
| | - Samuel Treviño
- Facultad de Química, Benemérita Universidad de Puebla, Puebla 72570, Mexico;
| | - Enrique Becerril-Villanueva
- Laboratorio de Psicoinmunología, Dirección de Neurociencias, Instituto Nacional de Psiquiatría “Ramón de la Fuente”, Mexico City 14370, Mexico;
| | - Carolina López-Rubalcava
- Departamento de Farmacobiología, Centro de Investigación y Estudios Avanzados del IPN, Mexico City 14330, Mexico;
| |
Collapse
|
29
|
Kim JD, Copperi F, Diano S. Microglia in Central Control of Metabolism. Physiology (Bethesda) 2024; 39:0. [PMID: 37962895 PMCID: PMC11283896 DOI: 10.1152/physiol.00021.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/12/2023] [Accepted: 11/12/2023] [Indexed: 11/15/2023] Open
Abstract
Beyond their role as brain immune cells, microglia act as metabolic sensors in response to changes in nutrient availability, thus playing a role in energy homeostasis. This review highlights the evidence and challenges of studying the role of microglia in metabolism regulation.
Collapse
Affiliation(s)
- Jung Dae Kim
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, New York, United States
| | - Francesca Copperi
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, New York, United States
| | - Sabrina Diano
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, New York, United States
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, New York, United States
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, New York, United States
| |
Collapse
|
30
|
Davis AB, Lloyd KR, Bollinger JL, Wohleb ES, Reyes TM. Adolescent high fat diet alters the transcriptional response of microglia in the prefrontal cortex in response to stressors in both male and female mice. Stress 2024; 27:2365864. [PMID: 38912878 PMCID: PMC11228993 DOI: 10.1080/10253890.2024.2365864] [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: 10/31/2023] [Accepted: 05/28/2024] [Indexed: 06/25/2024] Open
Abstract
Both obesity and high fat diets (HFD) have been associated with an increase in inflammatory gene expression within the brain. Microglia play an important role in early cortical development and may be responsive to HFD, particularly during sensitive windows, such as adolescence. We hypothesized that HFD during adolescence would increase proinflammatory gene expression in microglia at baseline and potentiate the microglial stress response. Two stressors were examined, a physiological stressor [lipopolysaccharide (LPS), IP] and a psychological stressor [15 min restraint (RST)]. From 3 to 7 weeks of age, male and female mice were fed standard control diet (SC, 20% energy from fat) or HFD (60% energy from fat). On P49, 1 h before sacrifice, mice were randomly assigned to either stressor exposure or control conditions. Microglia from the frontal cortex were enriched using a Percoll density gradient and isolated via fluorescence-activated cell sorting (FACS), followed by RNA expression analysis of 30 genes (27 target genes, three housekeeping genes) using Fluidigm, a medium throughput qPCR platform. We found that adolescent HFD induced sex-specific transcriptional response in cortical microglia, both at baseline and in response to a stressor. Contrary to our hypothesis, adolescent HFD did not potentiate the transcriptional response to stressors in males, but rather in some cases, resulted in a blunted or absent response to the stressor. This was most apparent in males treated with LPS. However, in females, potentiation of the LPS response was observed for select proinflammatory genes, including Tnfa and Socs3. Further, HFD increased the expression of Itgam, Ikbkb, and Apoe in cortical microglia of both sexes, while adrenergic receptor expression (Adrb1 and Adra2a) was changed in response to stressor exposure with no effect of diet. These data identify classes of genes that are uniquely affected by adolescent exposure to HFD and different stressor modalities in males and females.
Collapse
Affiliation(s)
- Alyshia B Davis
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Kelsey R Lloyd
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Justin L Bollinger
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Eric S Wohleb
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Teresa M Reyes
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
31
|
Yang K, Hou R, Zhao J, Wang X, Wei J, Pan X, Zhu X. Lifestyle effects on aging and CVD: A spotlight on the nutrient-sensing network. Ageing Res Rev 2023; 92:102121. [PMID: 37944707 DOI: 10.1016/j.arr.2023.102121] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/12/2023] [Accepted: 11/04/2023] [Indexed: 11/12/2023]
Abstract
Aging is widespread worldwide and a significant risk factor for cardiovascular disease (CVD). Mechanisms underlying aging have attracted considerable attention in recent years. Remarkably, aging and CVD overlap in numerous ways, with deregulated nutrient sensing as a common mechanism and lifestyle as a communal modifier. Interestingly, lifestyle triggers or suppresses multiple nutrient-related signaling pathways. In this review, we first present the composition of the nutrient-sensing network (NSN) and its metabolic impact on aging and CVD. Secondly, we review how risk factors closely associated with CVD, including adverse life states such as sedentary behavior, sleep disorders, high-fat diet, and psychosocial stress, contribute to aging and CVD, with a focus on the bridging role of the NSN. Finally, we focus on the positive effects of beneficial dietary interventions, specifically dietary restriction and the Mediterranean diet, on the regulation of nutrient metabolism and the delayed effects of aging and CVD that depend on the balance of the NSN. In summary, we expound on the interaction between lifestyle, NSN, aging, and CVD.
Collapse
Affiliation(s)
- Kaiying Yang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Rongyao Hou
- Department of Neurology, The Affiliated Hiser Hospital of Qingdao University, Qingdao 266000, China
| | - Jie Zhao
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xia Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jin Wei
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
32
|
Herrera G, Silvero C MJ, Becerra MC, Lasaga M, Scimonelli T. Modulatory role of α-MSH in hippocampal-dependent memory impairment, synaptic plasticity changes, oxidative stress, and astrocyte reactivity induced by short-term high-fat diet intake. Neuropharmacology 2023; 239:109688. [PMID: 37591460 DOI: 10.1016/j.neuropharm.2023.109688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/11/2023] [Accepted: 08/12/2023] [Indexed: 08/19/2023]
Abstract
High-fat diet (HFD) consumption is associated with cognitive deficits and neurodegenerative diseases. Since the hippocampus is extremely sensitive to pathophysiological changes, neuroinflammation and the concomitant oxidative stress induced by HFD can significantly interfere with hippocampal-dependent functions related to learning and memory. The neuropeptide alpha-melanocyte stimulating hormone (α-MSH) mediates neuroprotective actions in the central nervous system and can reverse the effects of neuroinflammation in cognitive functions that depend on the hippocampus. In this study, we used male Wistar rats to evaluate the effect of short-term HFD intake (5 days) plus a mild immune challenge, Lipopolysaccharide (LPS 10 μg/kg) on contextual fear, changes in structural plasticity, oxidative stress, and astrocyte reactivation in the hippocampus. We also determined the possible modulatory role of α-MSH. HFD consumption was associated with an increase in markers of oxidative stress (Advanced oxidation protein products and Malondialdehyde) in the dorsal hippocampus (DH). We also found changes in hippocampal structural synaptic plasticity, observing a decrease in total spine in the DH after HFD plus LPS. We observed astrocyte proliferation and a significant increase in the percentage of the area occupied by GFAP. Treatment with α-MSH (0.1 μg/0.25 μl) in the DH reversed the effect of short-term HFD plus LPS on contextual fear memory, oxidative stress, and spine density. α-MSH also reduced astrocyte proliferation. Our present results indicate that HFD consumption for a short period sensitizes the central nervous system (CNS) to a subsequent immune challenge and impairs contextual fear memory and that α-MSH could have a modulatory protective effect.
Collapse
Affiliation(s)
- Guadalupe Herrera
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET. Departamento de Farmacología Otto Orshinger, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.
| | - M Jazmín Silvero C
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica UNITEFA-CONICET. Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - M Cecilia Becerra
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica UNITEFA-CONICET. Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mercedes Lasaga
- Instituto de Investigaciones Biomédicas INBIOMED UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Teresa Scimonelli
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET. Departamento de Farmacología Otto Orshinger, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
33
|
Ince LM, Darling JS, Sanchez K, Bell KS, Melbourne JK, Davis LK, Nixon K, Gaudet AD, Fonken LK. Sex differences in microglia function in aged rats underlie vulnerability to cognitive decline. Brain Behav Immun 2023; 114:438-452. [PMID: 37709153 PMCID: PMC10790303 DOI: 10.1016/j.bbi.2023.09.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/07/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023] Open
Abstract
Aging is associated with a significant shift in immune system reactivity ("inflammaging"), as basal inflammation increases but protective responses to infection are compromised. The immune system exhibits considerable sex differences, which may influence the process of inflammaging, including immune cell activation and behavioral consequences of immune signaling (i.e., impaired memory). Here, we test the hypothesis that sex differences in immune aging may mediate sex differences in cognitive decline. Aged male and female rats received peripheral immune stimulation using lipopolysaccharide (LPS), then molecular, cellular, and behavioral outcomes were assessed. We observed that LPS-treated aged male rats showed cognitive impairment and increased neuroinflammatory responses relative to adult males. In contrast, aged female rats did not display these aging-related deficits. Using transcriptomic and flow cytometry analyses, we further observed significant age- and sex- dependent changes in immune cell populations in the brain parenchyma and meninges, indicating a broad shift in the neuroinflammatory environment that may potentiate these behavioral effects. Ovariectomized aged female rats were also resistant to inflammation-induced memory deficits, indicating that ovarian hormones are not required for the attenuated neuroinflammation in aged females. Overall, our results indicate that males have amplified inflammatory priming with age, which contributes to age-associated cognitive decline. Our findings highlight sexual dimorphism in mechanisms of aging, and suggest that sex is a crucial consideration for identifying therapies for aging and neuroinflammation.
Collapse
Affiliation(s)
- Louise M Ince
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Jeffrey S Darling
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Kevin Sanchez
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Kiersten S Bell
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Jennifer K Melbourne
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Lourdes K Davis
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA
| | - Kimberly Nixon
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Andrew D Gaudet
- Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA; Department of Psychology, University of Texas at Austin, Austin, TX 78712, USA; Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX 78712, USA
| | - Laura K Fonken
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
34
|
Zhang XL, Hollander CM, Khan MY, D'silva M, Ma H, Yang X, Bai R, Keeter CK, Galkina EV, Nadler JL, Stanton PK. Myeloid cell deficiency of the inflammatory transcription factor Stat4 protects long-term synaptic plasticity from the effects of a high-fat, high-cholesterol diet. Commun Biol 2023; 6:967. [PMID: 37783748 PMCID: PMC10545833 DOI: 10.1038/s42003-023-05304-0] [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: 07/04/2022] [Accepted: 08/30/2023] [Indexed: 10/04/2023] Open
Abstract
Neuroinflammation is associated with neurodegenerative diseases, including Alzheimer's and Parkinson's. The cytokine interleukin-12 activates signal transducer and activator of transcription 4 (Stat4), and consumption of a high-fat, high-cholesterol diet (HFD-C) and Stat4 activity are associated with inflammation, atherosclerosis, and a diabetic metabolic phenotype. In studies of in vitro hippocampal slices from control Stat4fl/flLdlr-/- mice fed a HFD-C diabetogenic diet, we show that Schaffer collateral-CA1 synapses exhibited larger reductions in activity-dependent, long-term potentiation (LTP) of synaptic transmission, compared to mice fed a standard diet. Glucose tolerance and insulin sensitivity shifts produced by HFD-C diet were reduced in Stat4ΔLysMLdlr-/- mice compared to Stat4fl/flLdlr-/- controls. Stat4ΔLysMLdlr-/- mice, which lack Stat4 under control of the LysMCre promoter, were resistant to HFD-C induced impairments in LTP. In contrast, Schaffer collateral-CA1 synapses in Stat4ΔLysMLdlr-/- mice fed the HFD-C diet showed larger LTP than control Stat4fl/flLdlr-/- mice. Expression of a number of neuroinflammatory and synaptic plasticity genes was reduced by HFD-C diet in control mice, and less affected by HFD-C diet in Stat4ΔLysMLdlr-/- mice. These data suggest that suppression of Stat4 activation may protect against effects of Western diet on cognition, type 2 diabetes, and reduce risk of Alzheimer's disease and other neurodegenerative disorders associated with neuroinflammation.
Collapse
Affiliation(s)
- Xiao-Lei Zhang
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Callie M Hollander
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Mohammad Yasir Khan
- Department of Pharmacology, New York Medical College, Valhalla, NY, 10595, USA
| | - Melinee D'silva
- Department of Pharmacology, New York Medical College, Valhalla, NY, 10595, USA
| | - Haoqin Ma
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Xinyuan Yang
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Robin Bai
- Department of Microbiology & Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Coles K Keeter
- Department of Microbiology & Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Elena V Galkina
- Department of Microbiology & Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Jerry L Nadler
- Department of Pharmacology, New York Medical College, Valhalla, NY, 10595, USA
- ACOS-Research VA Northern California Health Care System, Sacramento, CA, 95655, USA
| | - Patric K Stanton
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, 10595, USA.
| |
Collapse
|
35
|
Huerta-Canseco C, Caba M, Camacho-Morales A. Obesity-mediated Lipoinflammation Modulates Food Reward Responses. Neuroscience 2023; 529:37-53. [PMID: 37591331 DOI: 10.1016/j.neuroscience.2023.08.019] [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: 12/01/2022] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 08/19/2023]
Abstract
Accumulation of white adipose tissue (WAT) during obesity is associated with the development of chronic low-grade inflammation, a biological process known as lipoinflammation. Systemic and central lipoinflammation accumulates pro-inflammatory cytokines including IL-6, IL-1β and TNF-α in plasma and also in brain, disrupting neurometabolism and cognitive behavior. Obesity-mediated lipoinflammation has been reported in brain regions of the mesocorticolimbic reward circuit leading to alterations in the perception and consumption of ultra-processed foods. While still under investigation, lipoinflammation targets two major outcomes of the mesocorticolimbic circuit during food reward: perception and motivation ("Wanting") and the pleasurable feeling of feeding ("Liking"). This review will provide experimental and clinical evidence supporting the contribution of obesity- or overnutrition-related lipoinflammation affecting the mesocorticolimbic reward circuit and enhancing food reward responses. We will also address neuroanatomical targets of inflammatory profiles that modulate food reward responses during obesity and describe potential cellular and molecular mechanisms of overnutrition linked to addiction-like behavior favored by brain lipoinflammation.
Collapse
Affiliation(s)
| | - Mario Caba
- Centro de Investigaciones Biomédicas, Universidad Veracruzana, Xalapa, Mexico
| | - Alberto Camacho-Morales
- Department of Biochemistry, College of Medicine, Universidad Autónoma de Nuevo León, Monterrey, NL, Mexico; Neurometabolism Unit, Center for Research and Development in Health Sciences, Universidad Autónoma de Nuevo León, Monterrey, NL, Mexico.
| |
Collapse
|
36
|
Butler MJ, Mackey-Alfonso SE, Massa N, Baskin KK, Barrientos RM. Dietary fatty acids differentially impact phagocytosis, inflammatory gene expression, and mitochondrial respiration in microglial and neuronal cell models. Front Cell Neurosci 2023; 17:1227241. [PMID: 37636589 PMCID: PMC10448530 DOI: 10.3389/fncel.2023.1227241] [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: 05/31/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023] Open
Abstract
The consumption of diets high in saturated fatty acids and/or refined carbohydrates are associated with neuroinflammation, cognitive dysfunction, and neurodegenerative disease. In contrast, diets high in polyunsaturated fatty acids are associated with anti-inflammatory and neuroprotective effects. We have previously shown that high fat diet (HFD) consumption increases saturated fatty acids and decreases polyunsaturated fatty acids in the hippocampus. We have further shown that HFD elicits exaggerated neuroinflammation and reduced synaptic elements, and results in robust memory deficits in aged rats. Here, we examined the impact of palmitate, an abundant dietary saturated fat, on a variety of cellular responses in BV2 microglia and HippoE-14 neurons, and the extent to which the omega-3 fatty acid, docosahexaenoic acid (DHA), would buffer against these responses. Our data demonstrate that DHA pretreatment prevents or partially attenuates palmitate-induced alterations in proinflammatory, endoplasmic reticulum stress, and mitochondrial damage-associated gene expression in both cell types. Furthermore, we show that synaptoneurosomes isolated from aged, HFD-fed mice are engulfed by BV2 microglia at a faster rate than synaptoneurosomes isolated from aged, chow-fed mice, suggesting HFD alters signaling at synapses to hasten their engulfment by microglia. Consistent with this notion, we found modest increases in complement proteins and a decrease in CD47 protein expression on synaptoneurosomes isolated from the hippocampus of aged, HFD-fed mice. Interestingly, palmitate reduced BV2 microglial phagocytosis, but only of synaptoneurosomes isolated from chow-fed mice, an effect that was prevented by DHA pretreatment. Lastly, we measured the impact of palmitate and DHA on mitochondrial function in both microglial and neuronal cell models using the Seahorse XFe96 Analyzer. These data indicate that DHA pretreatment does not mitigate palmitate-induced reductions in mitochondrial respiration in BV2 microglia and HippoE-14 neurons, suggesting DHA may be acting downstream of mitochondrial function to exert its protective effects. Together, this study provides evidence that DHA can ameliorate the negative impact of palmitate on a variety of cellular functions in microglia- and neuron-like cells.
Collapse
Affiliation(s)
- Michael J. Butler
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, United States
| | - Sabrina E. Mackey-Alfonso
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
- Medical Scientist Training Program, The Ohio State University, Columbus, OH, United States
| | - Nashali Massa
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| | - Kedryn K. Baskin
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Ruth M. Barrientos
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, United States
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
- Chronic Brain Injury Program, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
37
|
González Olmo BM, Bettes MN, DeMarsh JW, Zhao F, Askwith C, Barrientos RM. Short-term high-fat diet consumption impairs synaptic plasticity in the aged hippocampus via IL-1 signaling. NPJ Sci Food 2023; 7:35. [PMID: 37460765 DOI: 10.1038/s41538-023-00211-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 07/06/2023] [Indexed: 07/20/2023] Open
Abstract
More Americans are consuming diets higher in saturated fats and refined sugars than ever before. These trends could have serious consequences for the older population because high-fat diet (HFD) consumption, known to induce neuroinflammation, has been shown to accelerate and aggravate memory declines. We have previously demonstrated that short-term HFD consumption, which does not evoke obesity-related comorbidities, produced profound impairments to hippocampal-dependent memory in aged rats. These impairments were precipitated by increases in proinflammatory cytokines, primarily interleukin-1 beta (IL-1β). Here, we explored the extent to which short-term HFD consumption disrupts hippocampal synaptic plasticity, as measured by long-term potentiation (LTP), in young adult and aged rats. We demonstrated that (1) HFD disrupted late-phase LTP in the hippocampus of aged, but not young adult rats, (2) HFD did not disrupt early-phase LTP, and (3) blockade of the IL-1 receptor rescued L-LTP in aged HFD-fed rats. These findings suggest that hippocampal memory impairments in aged rats following HFD consumption occur through the deterioration of synaptic plasticity and that IL-1β is a critical driver of that deterioration.
Collapse
Affiliation(s)
- Brigitte M González Olmo
- Department of Biomedical Education & Anatomy, Ohio State University, Columbus, OH, USA
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Menaz N Bettes
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - James W DeMarsh
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Fangli Zhao
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Candice Askwith
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA.
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA.
- Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, USA.
- Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
38
|
So SW, Fleming KM, Nixon JP, Butterick TA. Early Life Obesity Increases Neuroinflammation, Amyloid Beta Deposition, and Cognitive Decline in a Mouse Model of Alzheimer's Disease. Nutrients 2023; 15:nu15112494. [PMID: 37299457 DOI: 10.3390/nu15112494] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Obesity, a known risk factor of Alzheimer's disease (AD), increases the activation of microglia, leading to a proinflammatory phenotype. Our previous work shows that a high fat diet (HFD) can cause neuroinflammation and cognitive decline in mice. We hypothesized that proinflammatory activation of brain microglia in obesity exacerbates AD pathology and increases the accumulation of amyloid beta (Aβ) plaques. Presently, we tested cognitive function in 8-month-old male and female APP/PS1 mice fed a HFD, starting at 1.5 months of age. Locomotor activity, anxiety-like behavior, behavioral despair, and spatial memory were all assessed through behavioral tests. Microgliosis and Aβ deposition were measured in multiple brain regions through immunohistochemical analysis. Our results show that a HFD decreases locomotor activity, while increasing anxiety-like behavior and behavioral despair independent of genotype. A HFD led to increased memory deficits in both sexes, with HFD-fed APP/PS1 mice performing the worst out of all groups. Immunohistochemical analysis showed increased microgliosis in mice fed a HFD. This was accompanied by an increase in Aβ deposition in the HFD-fed APP/PS1 mice. Together, our results support that HFD-induced obesity exacerbates neuroinflammation and Aβ deposition in a young adult AD mouse model, leading to increased memory deficits and cognitive decline in both sexes.
Collapse
Affiliation(s)
- Simon W So
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN 55417, USA
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Kendra M Fleming
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN 55417, USA
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Joshua P Nixon
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN 55417, USA
- Department of Food Science and Nutrition, University of Minnesota Twin Cities, St. Paul, MN 55108, USA
- Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Tammy A Butterick
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN 55417, USA
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
- Department of Food Science and Nutrition, University of Minnesota Twin Cities, St. Paul, MN 55108, USA
| |
Collapse
|
39
|
Xiong W, Jiang X, He J, Liu X, Zhu Y, Liu B, Huang Y. Probiotic Fermentation of Kelp Enzymatic Hydrolysate Promoted its Anti-Aging Activity in D-Galactose-Induced Aging Mice by Modulating Gut Microbiota. Mol Nutr Food Res 2023; 67:e2200766. [PMID: 37005336 DOI: 10.1002/mnfr.202200766] [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: 11/05/2022] [Revised: 03/07/2023] [Indexed: 03/19/2023]
Abstract
SCOPE To investigate anti-aging effects of probiotic-fermented kelp enzymatic hydrolysate culture (KMF), probiotic-fermented kelp enzymatic hydrolysate supernatant (KMFS), and probiotic-fermented kelp enzymatic hydrolysate bacteria suspension (KMFP) in D-galactose-induced aging mice. METHODS AND RESULTS The study uses a probiotic-mixture of Lactobacillus reuteri, Pediococcus pentosaceus, and Lactobacillus acidophilus strains for kelp fermentation. KMF, KMFS, and KMFP prevent D-galactose-induced elevation of malondialdehyde levels in serum and brain tissue of aging mice, and they increase superoxide dismutase and catalase levels and total antioxidant capacity. Furthermore, they improve the cell structure of mouse brain, liver, and intestinal tissue. Compared with the model control group, the KMF, KMFS, and KMFP treatments regulate mRNA and protein levels of genes associated with aging, the concentrations of acetic acid, propionic acid, and butyric acid in the three treatment groups are more than 1.4-, 1.3-, and 1.2-fold increased, respectively. Furthermore, the treatments affect the gut microbiota community structures. CONCLUSIONS These results suggest that KMF, KMFS, and KMFP can modulate gut microbiota imbalances and positively affect aging-related genes to achieve anti-aging effects.
Collapse
Affiliation(s)
- Wenyu Xiong
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, China
- Engineering Research Center of Fujian and Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, 350002, China
- Fuzhou Ocean Research Institute Marine Food Research and Development Center, Fuzhou, 350002, China
| | - Xiaoqin Jiang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, China
- Engineering Research Center of Fujian and Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, 350002, China
- Fuzhou Ocean Research Institute Marine Food Research and Development Center, Fuzhou, 350002, China
| | - Junqiang He
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, China
- Engineering Research Center of Fujian and Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, 350002, China
- Fuzhou Ocean Research Institute Marine Food Research and Development Center, Fuzhou, 350002, China
| | - Xiaoyan Liu
- Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing, 100048, China
| | - Yuxian Zhu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, China
- Engineering Research Center of Fujian and Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, 350002, China
- Fuzhou Ocean Research Institute Marine Food Research and Development Center, Fuzhou, 350002, China
| | - Bin Liu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, China
- Engineering Research Center of Fujian and Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, 350002, China
- Fuzhou Ocean Research Institute Marine Food Research and Development Center, Fuzhou, 350002, China
| | - Ying Huang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, China
- Engineering Research Center of Fujian and Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, 350002, China
- Fuzhou Ocean Research Institute Marine Food Research and Development Center, Fuzhou, 350002, China
| |
Collapse
|
40
|
Chen X, Ma L, Zhao J, Pan X, Chen S. Association between cognitive impairment promoted by high-fat diet and increase in PTEN phosphorylation. Behav Brain Res 2023; 447:114421. [PMID: 37028516 DOI: 10.1016/j.bbr.2023.114421] [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: 01/30/2023] [Revised: 03/30/2023] [Accepted: 04/04/2023] [Indexed: 04/09/2023]
Abstract
The purpose of this study was to observe the changes in memory impairment and hippocampal phosphorylated protein levels in mice caused by obesity, and to explore the key phosphorylation modification proteins and pathways of memory impairment induced by high-fat diet. First, sixteen C57BL/6J mice were randomly divided into simple obese group (group H, n=8) and normal control group (group C, n=8). And at the end of the experiment, the cognitive function of the mice was assessed by Morris water maze and serological indexes were measured. Finally, phosphoproteomics was used to identify the differentially phosphorylted protein expression in the hippocampus of obese mice. Compared with group C, mice in group H had significantly decreased learning and memory abilities, and significantly increased body weight, blood glucose and lipid levels. The results of the phosphoproteomics analysis showed 442 up-regulated differentially phosphorylated proteins and 402 down-regulated differentially phosphorylated proteins. Further protein-protein interaction (PPI) analysis revealed pathway hub proteins, including β-actin (ACTB), Phosphatase and tensin homolog deleted on chromosome ten (PTEN), Phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1), mammalian target of rapamycin (mTOR), ribosomal protein 6 (RPS6), etc. Notably, the hub proteins PTEN, PIK3R1, and mTOR were jointly involved in the mTOR signaling pathway. Our study shows for the first time that a high-fat diet increases the phosphorylation of PTEN proteins, which may affect cognitive function.
Collapse
Affiliation(s)
- Xiaoyi Chen
- Graduate School of Hebei North University, Zhangjiakou, China; Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
| | - Liang Ma
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Jingyu Zhao
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China; Graduate School of North China University of Science and Technology, Tangshan, China
| | - Xiaoyu Pan
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
| | - Shuchun Chen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China.
| |
Collapse
|
41
|
Muscat SM, Butler MJ, Mackey-Alfonso SE, Barrientos RM. Young adult and aged female rats are vulnerable to amygdala-dependent, but not hippocampus-dependent, memory impairment following short-term high-fat diet. Brain Res Bull 2023; 195:145-156. [PMID: 36870621 PMCID: PMC10257807 DOI: 10.1016/j.brainresbull.2023.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/18/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023]
Abstract
Global populations are increasingly consuming diets high in saturated fats and refined carbohydrates, and such diets have been well-associated with heightened inflammation and neurological dysfunction. Notably, older individuals are particularly vulnerable to the impact of unhealthy diet on cognition, even after a single meal, and pre-clinical rodent studies have demonstrated that short-term consumption of high-fat diet (HFD) induces marked increases in neuroinflammation and cognitive impairment. Unfortunately though, to date, most studies on the topic of nutrition and cognition, especially in aging, have been performed only in male rodents. This is especially concerning given that older females are more vulnerable to develop certain memory deficits and/or severe memory-related pathologies than males. Thus, the aim of the present study was to determine the extent to which short-term HFD consumption impacts memory function and neuroinflammation in female rats. Young adult (3 months) and aged (20-22 months) female rats were fed HFD for 3 days. Using contextual fear conditioning, we found that HFD had no effect on long-term contextual memory (hippocampus-dependent) at either age, but impaired long-term auditory-cued memory (amygdala-dependent) regardless of age. Gene expression of Il-1β was markedly dysregulated in the amygdala, but not hippocampus, of both young and aged rats after 3 days of HFD. Interestingly, modulation of IL-1 signaling via central administration of the IL-1 receptor antagonist (which we have previously demonstrated to be protective in males) had no impact on memory function following the HFD in females. Investigation of the memory-associated gene Pacap and its receptor Pac1r revealed differential effects of HFD on their expression in the hippocampus and amygdala. Specifically, HFD induced increased expression of Pacap and Pac1r in the hippocampus, whereas decreased Pacap was observed in the amygdala. Collectively, these data suggest that both young adult and aged female rats are vulnerable to amygdala-dependent (but not hippocampus-dependent) memory impairments following short-term HFD consumption, and identify potential mechanisms related to IL-1β and PACAP signaling in these differential effects. Notably, these findings are strikingly different than those previously reported in male rats using the same diet regimen and behavioral paradigms, and highlight the importance of examining potential sex differences in the context of neuroimmune-associated cognitive dysfunction.
Collapse
Affiliation(s)
- Stephanie M Muscat
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Michael J Butler
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Sabrina E Mackey-Alfonso
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Medical Scientist Training Program, The Ohio State University, Columbus, OH, USA
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
42
|
Nadjar A. Discovery of a new type of neuroimmune interaction in the aged brain's response to energy surfeit. Brain Behav Immun 2023; 110:288-289. [PMID: 36914015 DOI: 10.1016/j.bbi.2023.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 03/08/2023] [Indexed: 03/15/2023] Open
Affiliation(s)
- Agnes Nadjar
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France; Institut Universitaire de France (IUF), France.
| |
Collapse
|
43
|
Dumas JA, Bunn JY, LaMantia MA, McIsaac C, Senft Miller A, Nop O, Testo A, Soares BP, Mank MM, Poynter ME, Lawrence Kien C. Alteration of brain function and systemic inflammatory tone in older adults by decreasing the dietary palmitic acid intake. AGING BRAIN 2023; 3:100072. [PMID: 37408793 PMCID: PMC10318304 DOI: 10.1016/j.nbas.2023.100072] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/12/2023] Open
Abstract
Prior studies in younger adults showed that reducing the normally high intake of the saturated fatty acid, palmitic acid (PA), in the North American diet by replacing it with the monounsaturated fatty acid, oleic acid (OA), decreased blood concentrations and secretion by peripheral blood mononuclear cells (PBMCs) of interleukin (IL)-1β and IL-6 and changed brain activation in regions of the working memory network. We examined the effects of these fatty acid manipulations in the diet of older adults. Ten subjects, aged 65-75 years, participated in a randomized, cross-over trial comparing 1-week high PA versus low PA/high OA diets. We evaluated functional magnetic resonance imaging (fMRI) using an N-back test of working memory and a resting state scan, cytokine secretion by lipopolysaccharide (LPS)-stimulated PBMCs, and plasma cytokine concentrations. During the low PA compared to the high PA diet, we observed increased activation for the 2-back minus 0-back conditions in the right dorsolateral prefrontal cortex (Broadman Area (BA) 9; p < 0.005), but the effect of diet on working memory performance was not significant (p = 0.09). We observed increased connectivity between anterior regions of the salience network during the low PA/high OA diet (p < 0.001). The concentrations of IL-1β (p = 0.026), IL-8 (p = 0.013), and IL-6 (p = 0.009) in conditioned media from LPS-stimulated PBMCs were lower during the low PA/high OA diet. This study suggests that lowering the dietary intake of PA down-regulated pro-inflammatory cytokine secretion and altered working memory, task-based activation and resting state functional connectivity in older adults.
Collapse
Affiliation(s)
- Julie A. Dumas
- Department of Psychiatry, Larner College of Medicine, The University of Vermont, Burlington, VT, USA
| | - Janice Y. Bunn
- Department of Medical Biostatistics, Larner College of Medicine, The University of Vermont, Burlington, VT, USA
| | - Michael A. LaMantia
- Department of Medicine, Larner College of Medicine, The University of Vermont, Burlington, VT, USA
| | - Catherine McIsaac
- Clinical Research Center, Larner College of Medicine, The University of Vermont, Burlington, VT, USA
| | - Anna Senft Miller
- Department of Psychiatry, Larner College of Medicine, The University of Vermont, Burlington, VT, USA
| | - Olivia Nop
- Department of Psychiatry, Larner College of Medicine, The University of Vermont, Burlington, VT, USA
| | - Abigail Testo
- Department of Psychiatry, Larner College of Medicine, The University of Vermont, Burlington, VT, USA
| | - Bruno P. Soares
- Department of Radiology, Larner College of Medicine, The University of Vermont, Burlington, VT, USA
| | - Madeleine M. Mank
- Department of Medicine, Larner College of Medicine, The University of Vermont, Burlington, VT, USA
| | - Matthew E. Poynter
- Department of Medicine, Larner College of Medicine, The University of Vermont, Burlington, VT, USA
| | - C. Lawrence Kien
- Department of Medicine, Larner College of Medicine, The University of Vermont, Burlington, VT, USA
- Department of Pediatrics, Larner College of Medicine, The University of Vermont, Burlington, VT, USA
| |
Collapse
|
44
|
Butler MJ, Sengupta S, Muscat SM, Amici SA, Biltz RG, Deems NP, Dravid P, Mackey-Alfonso S, Ijaz H, Bettes MN, Godbout JP, Kapoor A, Guerau-de-Arellano M, Barrientos RM. CD8 + T cells contribute to diet-induced memory deficits in aged male rats. Brain Behav Immun 2023; 109:235-250. [PMID: 36764399 PMCID: PMC10124165 DOI: 10.1016/j.bbi.2023.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/27/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
We have previously shown that short-term (3-day) high fat diet (HFD) consumption induces a neuroinflammatory response and subsequent impairment of long-term memory in aged, but not young adult, male rats. However, the immune cell phenotypes driving this proinflammatory response are not well understood. Previously, we showed that microglia isolated from young and aged rats fed a HFD express similar levels of priming and proinflammatory transcripts, suggesting that additional factors may drive the exaggerated neuroinflammatory response selectively observed in aged HFD-fed rats. It is established that T cells infiltrate both the young and especially the aged central nervous system (CNS) and contribute to immune surveillance of the parenchyma. Thus, we investigated the modulating role of short-term HFD on T cell presence in the CNS in aged rats using bulk RNA sequencing and flow cytometry. RNA sequencing results indicate that aging and HFD altered the expression of genes and signaling pathways associated with T cell signaling, immune cell trafficking, and neuroinflammation. Moreover, flow cytometry data showed that aging alone increased CD4+ and CD8+ T cell presence in the brain and that CD8+, but not CD4+, T cells were further increased in aged rats fed a HFD. Based on these data, we selectively depleted circulating CD8+ T cells via an intravenous injection of an anti-CD8 antibody in aged rats prior to 3 days of HFD to infer the functional role these cells may be playing in long-term memory and neuroinflammation. Results indicate that peripheral depletion of CD8+ T cells lowered hippocampal cytokine levels and prevented the HFD-induced i) increase in brain CD8+ T cells, ii) memory impairment, and iii) alterations in pre- and post-synaptic structures in the hippocampus and amygdala. Together, these data indicate a substantial role for CD8+ T cells in mediating diet-induced memory impairments in aged male rats.
Collapse
Affiliation(s)
- Michael J Butler
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA.
| | - Shouvonik Sengupta
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Stephanie M Muscat
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Stephanie A Amici
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH, USA
| | - Rebecca G Biltz
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Nicholas P Deems
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Piyush Dravid
- College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Sabrina Mackey-Alfonso
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Haanya Ijaz
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Menaz N Bettes
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Jonathan P Godbout
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH 43210, USA
| | - Amit Kapoor
- College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Mireia Guerau-de-Arellano
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH, USA
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
45
|
Hamilton J, Nguyen C, McAvoy M, Roeder N, Richardson B, Quattrin T, Hajnal A, Thanos PK. Calorie restriction, but not Roux-en-Y gastric bypass surgery, increases [ 3 H] PK11195 binding in a rat model of obesity. Synapse 2023; 77:e22258. [PMID: 36352528 DOI: 10.1002/syn.22258] [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: 03/28/2022] [Revised: 10/05/2022] [Accepted: 10/09/2022] [Indexed: 11/11/2022]
Abstract
Roux-en-Y gastric bypass surgery (RYGB) remains an effective weight-loss method used to treat obesity. While it is successful in combating obesity, there are many lingering questions related to the changes in the brain following RYGB surgery, one of them being its effects on neuroinflammation. While it is known that chronic high-fat diet (HFD) contributes to obesity and neuroinflammation, it remains to be understood whether bariatric surgery can ameliorate diet-induced inflammatory responses. To examine this, rats were assigned to either a normal diet (ND) or a HFD for 8 weeks. Rats fed a HFD were split into the following groups: sham surgery with ad libitum access to HFD (sham-HF); sham surgery with calorie-restricted HFD (sham-FR); RYGB surgery with ad libitum access to HFD (RYGB). Following sham or RYGB surgeries, rats were maintained on their diets for 9 weeks before being euthanized. [3 H] PK11195 autoradiography was then performed on fresh-frozen brain tissue in order to measure activated microglia. Sham-FR rats showed increased [3 H] PK11195 binding in the amygdala (63%), perirhinal (60%), and ectorhinal cortex (53%) compared with the ND rats. Obese rats who had the RYGB surgery did not show this increased inflammatory effect. Since the sham-FR and RYGB rats were fed the same amount of HFD, the surgery itself seems responsible for this attenuation in [3 H] PK11195 binding. We speculate that calorie restriction following obese conditions may be seen as a stressor and contribute to inflammation in the brain. Further research is needed to verify this mechanism.
Collapse
Affiliation(s)
- John Hamilton
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Cynthia Nguyen
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Margaret McAvoy
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Nicole Roeder
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA.,Department of Psychology, University at Buffalo, Buffalo, New York, USA
| | - Brittany Richardson
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA.,Department of Psychology, University at Buffalo, Buffalo, New York, USA
| | - Teresa Quattrin
- Department of Pediatrics, University at Buffalo, UBMD Pediatrics, JR Oishei Children's Hospital, Buffalo, New York, USA
| | - Andras Hajnal
- Department of Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Panayotis K Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA.,Department of Psychology, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
46
|
Long-Term Ingestion of Sicilian Black Bee Chestnut Honey and/or D-Limonene Counteracts Brain Damage Induced by High Fat-Diet in Obese Mice. Int J Mol Sci 2023; 24:ijms24043467. [PMID: 36834882 PMCID: PMC9966634 DOI: 10.3390/ijms24043467] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/25/2023] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
Obesity is linked to neurodegeneration, which is mainly caused by inflammation and oxidative stress. We analyzed whether the long-term intake of honey and/or D-limonene, which are known for their antioxidant and anti-inflammatory actions, when ingested separately or in combination, can counteract the neurodegeneration occurring in high fat diet (HFD)-induced obesity. After 10 weeks of HFD, mice were divided into: HFD-, HFD + honey (HFD-H)-, HFD + D-limonene (HFD-L)-, HFD + honey + D-limonene (HFD-H + L)-fed groups, for another 10 weeks. Another group was fed a standard diet (STD). We analyzed the brain neurodegeneration, inflammation, oxidative stress, and gene expression of Alzheimer's disease (AD) markers. The HFD animals showed higher neuronal apoptosis, upregulation of pro-apoptotic genes Fas-L, Bim P27 and downregulation of anti-apoptotic factors BDNF and BCL2; increased gene expression of the pro-inflammatory IL-1β, IL-6 and TNF-α and elevated oxidative stress markers COX-2, iNOS, ROS and nitrite. The honey and D-limonene intake counteracted these alterations; however, they did so in a stronger manner when in combination. Genes involved in amyloid plaque processing (APP and TAU), synaptic function (Ache) and AD-related hyperphosphorylation were higher in HFD brains, and significantly downregulated in HFD-H, HFD-L and HFD-H + L. These results suggest that honey and limonene ingestion counteract obesity-related neurodegeneration and that joint consumption is more efficacious than a single administration.
Collapse
|
47
|
Muscat SM, Deems NP, Butler MJ, Scaria EA, Bettes MN, Cleary SP, Bockbrader RH, Maier SF, Barrientos RM. Selective TLR4 Antagonism Prevents and Reverses Morphine-Induced Persistent Postoperative Cognitive Dysfunction, Dysregulation of Synaptic Elements, and Impaired BDNF Signaling in Aged Male Rats. J Neurosci 2023; 43:155-172. [PMID: 36384680 PMCID: PMC9838714 DOI: 10.1523/jneurosci.1151-22.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 11/07/2022] [Accepted: 11/10/2023] [Indexed: 11/18/2022] Open
Abstract
Perioperative neurocognitive disorders (PNDs) are characterized by confusion, difficulty with executive function, and episodic memory impairment in the hours to months following a surgical procedure. Postoperative cognitive dysfunction (POCD) represents such impairments that last beyond 30 d postsurgery and is associated with increased risk of comorbidities, progression to dementia, and higher mortality. While it is clear that neuroinflammation plays a key role in PND development, what factors underlie shorter self-resolving versus persistent PNDs remains unclear. We have previously shown that postoperative morphine treatment extends POCD from 4 d (without morphine) to at least 8 weeks (with morphine) in aged male rats, and that this effect is likely dependent on the proinflammatory capabilities of morphine via activation of toll-like receptor 4 (TLR4). Here, we extend these findings to show that TLR4 blockade, using the selective TLR4 antagonist lipopolysaccharide from the bacterium Rhodobacter sphaeroides (LPS-RS Ultrapure), ameliorates morphine-induced POCD in aged male rats. Using either a single central preoperative treatment or a 1 week postoperative central treatment regimen, we demonstrate that TLR4 antagonism (1) prevents and reverses the long-term memory impairment associated with surgery and morphine treatment, (2) ameliorates morphine-induced dysregulation of the postsynaptic proteins postsynaptic density 95 and synaptopodin, (3) mitigates reductions in mature BDNF, and (4) prevents decreased activation of the BDNF receptor TrkB (tropomyosin-related kinase B), all at 4 weeks postsurgery. We also reveal that LPS-RS Ultrapure likely exerts its beneficial effects by preventing endogenous danger signal HMGB1 (high-mobility group box 1) from activating TLR4, rather than by blocking continuous activation by morphine or its metabolites. These findings suggest TLR4 as a promising therapeutic target to prevent or treat PNDs.SIGNIFICANCE STATEMENT With humans living longer than ever, it is crucial that we identify mechanisms that contribute to aging-related vulnerability to cognitive impairment. Here, we show that the innate immune receptor toll-like receptor 4 (TLR4) is a key mediator of cognitive dysfunction in aged rodents following surgery and postoperative morphine treatment. Inhibition of TLR4 both prevented and reversed surgery plus morphine-associated memory impairment, dysregulation of synaptic elements, and reduced BDNF signaling. Together, these findings implicate TLR4 in the development of postoperative cognitive dysfunction, providing mechanistic insight and novel therapeutic targets for the treatment of cognitive impairments following immune challenges such as surgery in older individuals.
Collapse
Affiliation(s)
- Stephanie M Muscat
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio 43210
| | - Nicholas P Deems
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
- Neuroscience Graduate Program, The Ohio State University, Columbus, Ohio 43210
| | - Michael J Butler
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
| | - Emmanuel A Scaria
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
| | - Menaz N Bettes
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
| | - Sean P Cleary
- Campus Chemical Instrumentation Center, The Ohio State University, Columbus, Ohio 43210
| | - Ross H Bockbrader
- Pharmaceutical Sciences Graduate Program, Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, Columbus, Ohio 43210
| | - Steven F Maier
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado Boulder, Boulder, Colorado 80309
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio 43210
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, Ohio 43210
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
- Chronic Brain Injury Program, The Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
48
|
Wei W, Lin Z, Xu P, Lv X, Lin L, Li Y, Zhou Y, Lu T, Xue X. Diet Control and Swimming Exercise Ameliorate HFD-Induced Cognitive Impairment Related to the SIRT1-NF- κB/PGC-1 α Pathways in ApoE-/- Mice. Neural Plast 2023; 2023:9206875. [PMID: 36999158 PMCID: PMC10049848 DOI: 10.1155/2023/9206875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/04/2023] [Accepted: 03/07/2023] [Indexed: 04/01/2023] Open
Abstract
High-fat diet- (HFD-) induced neuroinflammation may ultimately lead to an increased risk of cognitive impairment. Here, we evaluate the effects of diet control and swimming or both on the prevention of cognitive impairment by enhancing SIRT1 activity. Twenty-week-old ApoE-/- mice were fed a HFD for 8 weeks and then were treated with diet control and/or swimming for 8 weeks. Cognitive function was assessed using the novel object recognition test (NORT) and Y-maze test. The expression of sirtuin-1 (SIRT1), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), brain-derived neurotrophic factor (BDNF), nuclear factor kappa B p65 (NF-κB p65), interleukin-1β (IL-1β), and tumour necrosis factor-α (TNF-α) in the hippocampus was measured by western blotting. The levels of fractional anisotropy (FA), N-acetylaspartate (NAA)/creatine (Cr) ratio, choline (Cho)/Cr ratio, and myo-inositol (MI)/Cr ratio in the hippocampus were evaluated by diffusion tensor imaging (DTI) and magnetic resonance spectroscopy (MRS) using 7.0-T magnetic resonance imaging (MRI). Our results showed that cognitive dysfunction and hippocampal neuroinflammation appeared to be remarkably observed in apolipoprotein E (ApoE)-/- mice fed with HFD. Diet control plus swimming significantly reversed HFD-induced cognitive decline, reduced the time spent exploring the novel object, and ameliorated spontaneous alternation in the Y-maze test. Compared with the HFD group, ApoE-/- mice fed diet control and/or subjected to swimming had an increase in FA, NAA/Cr, and Cho/Cr; a drop in MI/Cr; elevated expression levels of SIRT1, PGC-1α, and BDNF; and inhibited production of proinflammatory cytokines, including NF-κB p65, IL-1β, and TNF-α. SIRT1, an NAD+-dependent class III histone enzyme, deacetylases and regulates the activity of PGC-1α and NF-κB. These data indicated that diet control and/or swimming ameliorate cognitive deficits through the inhibitory effect of neuroinflammation via SIRT1-mediated pathways, strongly suggesting that swimming and/or diet control could be potentially effective nonpharmacological treatments for cognitive impairment.
Collapse
Affiliation(s)
- Wei Wei
- 1The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Zhicheng Lin
- 1The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - PeiTao Xu
- 1The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xinru Lv
- 2College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Libin Lin
- 2College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yongxu Li
- 2College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yangjie Zhou
- 2College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Taotao Lu
- 2College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xiehua Xue
- 1The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- 3Fujian Provincial Rehabilitation Industrial Institution, Fujian Provincial Key Laboratory of Rehabilitation Technology, Fujian Key Laboratory of Cognitive Rehabilitation, Fuzhou, China
| |
Collapse
|
49
|
ERTAL E, ÖZKAYA V. Düşük veya Yüksek Karbonhidratlı Diyetlerin Beyin, Beyin-Bağırsak Aksı ve Bilişsel İşlevler Üzerine Etkisi. İSTANBUL GELIŞIM ÜNIVERSITESI SAĞLIK BILIMLERI DERGISI 2022. [DOI: 10.38079/igusabder.1140592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
İntestinal mikrobiyota sağlığın korunmasında anahtar bir rol oynamaktadır. Mikrobiyota üzerine önemli etkileri olan beslenme, beyin-bağırsak aksındaki bozuklukları hafifletmek, nöroinflamasyonu ve bilişsel bozulmayı iyileştirmek için büyük önem taşımaktadır. Bağırsak bakterileri, diyetle alınan besin ögelerini kullanarak çeşitli metabolitleri (örn., kısa zincirli yağ asitleri, amino asitler, vitaminler) üretebilme yeteneğine sahiptir. Üretilen bu metabolitler, periferik sinir sistemi, enteroendokrin hücreler ve merkezi sinir sistemine sinyal gönderen immün hücreler aracılığıyla beyin fonksiyonlarını ve bilişsel davranış değişikliğini etkilemektedir. Karbonhidratlar, çoğu durumda intestinal mikrobiyota tarafından substrat olarak kullanılmakta ve fermente edilmektedir. Karbonhidratların bu etkileri kimyasal yapılarına, sindirilmeden kolona ulaşıp ulaşamamalarına ve konağın karbonhidratı enerji kaynağı olarak kullanabilme yeteneğine bağlıdır. Karbonhidratın türü ve miktarı mikrobiyota, beyin bağırsak aksı ve bilişsel işlevlerdeki etkiyi belirleyen ana faktörlerden biridir. Bu derlemede, düşük veya yüksek karbonhidrat içeren diyetlerin beyin-bağırsak aksı ve bilişsel fonksiyonlara olan etkilerinin güncel literatür verileri ışığında değerlendirilmesi amaçlanmıştır.
Collapse
Affiliation(s)
- Ezgi ERTAL
- BİRUNİ ÜNİVERSİTESİ, SAĞLIK BİLİMLERİ FAKÜLTESİ, BESLENME VE DİYETETİK BÖLÜMÜ
| | - Volkan ÖZKAYA
- İSTANBUL MEDİPOL ÜNİVERSİTESİ, SAĞLIK BİLİMLERİ FAKÜLTESİ, BESLENME VE DİYETETİK BÖLÜMÜ
| |
Collapse
|
50
|
Henn RE, Elzinga SE, Glass E, Parent R, Guo K, Allouch AM, Mendelson FE, Hayes J, Webber-Davis I, Murphy GG, Hur J, Feldman EL. Obesity-induced neuroinflammation and cognitive impairment in young adult versus middle-aged mice. Immun Ageing 2022; 19:67. [PMID: 36550567 PMCID: PMC9773607 DOI: 10.1186/s12979-022-00323-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Obesity rates are increasing worldwide. Obesity leads to many complications, including predisposing individuals to the development of cognitive impairment as they age. Immune dysregulation, including inflammaging (e.g., increased circulating cytokines) and immunosenescence (declining immune system function), commonly occur in obesity and aging and may impact cognitive impairment. As such, immune system changes across the lifespan may impact the effects of obesity on neuroinflammation and associated cognitive impairment. However, the role of age in obesity-induced neuroinflammation and cognitive impairment is unclear. To further define this putative relationship, the current study examined metabolic and inflammatory profiles, along with cognitive changes using a high-fat diet (HFD) mouse model of obesity. RESULTS First, HFD promoted age-related changes in hippocampal gene expression. Given this early HFD-induced aging phenotype, we fed HFD to young adult and middle-aged mice to determine the effect of age on inflammatory responses, metabolic profile, and cognitive function. As anticipated, HFD caused a dysmetabolic phenotype in both age groups. However, older age exacerbated HFD cognitive and neuroinflammatory changes, with a bi-directional regulation of hippocampal inflammatory gene expression. CONCLUSIONS Collectively, these data indicate that HFD promotes an early aging phenotype in the brain, which is suggestive of inflammaging and immunosenescence. Furthermore, age significantly compounded the impact of HFD on cognitive outcomes and on the regulation of neuroinflammatory programs in the brain.
Collapse
Affiliation(s)
- Rosemary E Henn
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sarah E Elzinga
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Emily Glass
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Molecular and Integrative Physiology, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Rachel Parent
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Molecular and Integrative Physiology, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kai Guo
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, 58202, USA
| | - Adam M Allouch
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Faye E Mendelson
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, 48109, USA
| | - John Hayes
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ian Webber-Davis
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Geoffery G Murphy
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Molecular and Integrative Physiology, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, 58202, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA.
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|