1
|
Traetta ME, Vecchiarelli HA, Tremblay MÈ. Fundamental Neurochemistry Review: Lipids across microglial states. J Neurochem 2025; 169:e16259. [PMID: 39696753 DOI: 10.1111/jnc.16259] [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: 08/08/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 12/20/2024]
Abstract
The capacity of immune cells to alter their function based on their metabolism is the basis of the emerging field of immunometabolism. Microglia are the resident innate immune cells of the central nervous system, and it is a current focus of the field to investigate how alterations in their metabolism impact these cells. Microglia have the ability to utilize lipids, such as fatty acids, as energy sources, but also alterations in lipids can impact microglial form and function. Recent studies highlighting different microglial states and transcriptional signatures have highlighted modifications in lipid processing as defining these states. This review highlights these recent studies and uses these altered pathways to discuss the current understanding of lipid biology in microglia. The studies highlighted here review how lipids may alter microglial phagocytic functioning or alter their pro- and anti-inflammatory balance. These studies provide a foundation by which lipid supplementation or diet alterations could influence microglial states and function. Furthermore, targets modulating microglial lipid metabolism may provide new treatment avenues.
Collapse
Affiliation(s)
- Marianela E Traetta
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Haley A Vecchiarelli
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, British Columbia, Canada
- Institute for Aging and Lifelong Health (IALH), University of Victoria, Victoria, British Columbia, Canada
- Département de médecine moléculaire, Université Laval, Québec City, Quebec, Canada
- Axe neurosciences, Centre de recherche du CHU de Québec, Université Laval, Québec City, Quebec, Canada
- Neurology and Neurosurgery Department, McGill University, Montréal, Quebec, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
2
|
Chen TB, Yang CC, Tsai IJ, Yang HW, Hsu YC, Chang CM, Yang CP. Neuroimmunological effects of omega-3 fatty acids on migraine: a review. Front Neurol 2024; 15:1366372. [PMID: 38770523 PMCID: PMC11103013 DOI: 10.3389/fneur.2024.1366372] [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: 01/08/2024] [Accepted: 04/12/2024] [Indexed: 05/22/2024] Open
Abstract
Migraine is a highly prevalent disease worldwide, imposing enormous clinical and economic burdens on individuals and societies. Current treatments exhibit limited efficacy and acceptability, highlighting the need for more effective and safety prophylactic approaches, including the use of nutraceuticals for migraine treatment. Migraine involves interactions within the central and peripheral nervous systems, with significant activation and sensitization of the trigeminovascular system (TVS) in pain generation and transmission. The condition is influenced by genetic predispositions and environmental factors, leading to altered sensory processing. The neuroinflammatory response is increasingly recognized as a key event underpinning the pathophysiology of migraine, involving a complex neuro-glio-vascular interplay. This interplay is partially mediated by neuropeptides such as calcitonin gene receptor peptide (CGRP), pituitary adenylate cyclase activating polypeptide (PACAP) and/or cortical spreading depression (CSD) and involves oxidative stress, mitochondrial dysfunction, nucleotide-binding domain-like receptor family pyrin domain containing-3 (NLRP3) inflammasome formation, activated microglia, and reactive astrocytes. Omega-3 polyunsaturated fatty acids (PUFAs), particularly eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), crucial for the nervous system, mediate various physiological functions. Omega-3 PUFAs offer cardiovascular, neurological, and psychiatric benefits due to their potent anti-inflammatory, anti-nociceptive, antioxidant, and neuromodulatory properties, which modulate neuroinflammation, neurogenic inflammation, pain transmission, enhance mitochondrial stability, and mood regulation. Moreover, specialized pro-resolving mediators (SPMs), a class of PUFA-derived lipid mediators, regulate pro-inflammatory and resolution pathways, playing significant anti-inflammatory and neurological roles, which in turn may be beneficial in alleviating the symptomatology of migraine. Omega-3 PUFAs impact various neurobiological pathways and have demonstrated a lack of major adverse events, underscoring their multifaceted approach and safety in migraine management. Although not all omega-3 PUFAs trials have shown beneficial in reducing the symptomatology of migraine, further research is needed to fully establish their clinical efficacy and understand the precise molecular mechanisms underlying the effects of omega-3 PUFAs and PUFA-derived lipid mediators, SPMs on migraine pathophysiology and progression. This review highlights their potential in modulating brain functions, such as neuroimmunological effects, and suggests their promise as candidates for effective migraine prophylaxis.
Collapse
Affiliation(s)
- Ting-Bin Chen
- Department of Neurology, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Cheng-Chia Yang
- Department of Healthcare Administration, Asia University, Taichung, Taiwan
| | - I-Ju Tsai
- Department of Neurology, Kuang Tien General Hospital, Taichung, Taiwan
- Department of Medical Research, Kuang Tien General Hospital, Taichung, Taiwan
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan
| | - Hao-Wen Yang
- Department of Family Medicine, Kuang Tien General Hospital, Taichung, Taiwan
| | - Yung-Chu Hsu
- Division of Neurology, Department of Internal Medicine, Ditmanson Medical Foundation ChiaYi Chistian Hospital, Chiayi, Taiwan
| | - Ching-Mao Chang
- Center for Traditional Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Pai Yang
- Department of Neurology, Kuang Tien General Hospital, Taichung, Taiwan
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
3
|
Baqueiro MDN, Simino LADP, Costa JP, Panzarin C, Reginato A, Torsoni MA, Ignácio-Souza L, Milanski M, Ross MG, Coca KP, Desai M, Torsoni AS. Sex-Dependent Variations in Hypothalamic Fatty Acid Profile and Neuropeptides in Offspring Exposed to Maternal Obesity and High-Fat Diet. Nutrients 2024; 16:340. [PMID: 38337626 PMCID: PMC10857148 DOI: 10.3390/nu16030340] [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: 11/28/2023] [Revised: 01/03/2024] [Accepted: 01/17/2024] [Indexed: 02/12/2024] Open
Abstract
Maternal obesity and/or high-fat diet (HF) consumption can disrupt appetite regulation in their offspring, contributing to transgenerational obesity and metabolic diseases. As fatty acids (FAs) play a role in appetite regulation, we investigated the maternal and fetal levels of FAs as potential contributors to programmed hyperphagia observed in the offspring of obese dams. Female mice were fed either a control diet (CT) or HF prior to mating, and fetal and maternal blood and tissues were collected at 19 days of gestation. Elevated levels of linoleic acid were observed in the serum of HF dams as well as in the serum of their fetuses. An increased concentration of eicosadienoic acid was also detected in the hypothalamus of female HF-O fetuses. HF-O male fetuses showed increased hypothalamic neuropeptide Y (Npy) gene expression, while HF-O female fetuses showed decreased hypothalamic pro-opiomelanocortin (POMC) protein content. Both male and female fetuses exhibited reduced hypothalamic neurogenin 3 (NGN-3) gene expression. In vitro experiments confirmed that LA contributed to the decreased gene expression of Pomc and Ngn-3 in neuronal cells. During lactation, HF female offspring consumed more milk and had a higher body weight compared to CT. In summary, this study demonstrated that exposure to HF prior to and during gestation alters the FA composition in maternal serum and fetal serum and hypothalamus, particularly increasing n-6, which may play a role in the switch from POMC to NPY neurons, leading to increased weight gain in the offspring during lactation.
Collapse
Affiliation(s)
- Mayara da Nóbrega Baqueiro
- Faculdade de Ciências Aplicadas, Universidade de Campinas, UNICAMP, Limeira 13484-350, São Paulo, Brazil; (M.d.N.B.); (L.A.d.P.S.); (J.P.C.); (C.P.); (A.R.); (M.A.T.); (L.I.-S.); (M.M.)
| | - Laís Angélica de Paula Simino
- Faculdade de Ciências Aplicadas, Universidade de Campinas, UNICAMP, Limeira 13484-350, São Paulo, Brazil; (M.d.N.B.); (L.A.d.P.S.); (J.P.C.); (C.P.); (A.R.); (M.A.T.); (L.I.-S.); (M.M.)
| | - João Paulo Costa
- Faculdade de Ciências Aplicadas, Universidade de Campinas, UNICAMP, Limeira 13484-350, São Paulo, Brazil; (M.d.N.B.); (L.A.d.P.S.); (J.P.C.); (C.P.); (A.R.); (M.A.T.); (L.I.-S.); (M.M.)
| | - Carolina Panzarin
- Faculdade de Ciências Aplicadas, Universidade de Campinas, UNICAMP, Limeira 13484-350, São Paulo, Brazil; (M.d.N.B.); (L.A.d.P.S.); (J.P.C.); (C.P.); (A.R.); (M.A.T.); (L.I.-S.); (M.M.)
| | - Andressa Reginato
- Faculdade de Ciências Aplicadas, Universidade de Campinas, UNICAMP, Limeira 13484-350, São Paulo, Brazil; (M.d.N.B.); (L.A.d.P.S.); (J.P.C.); (C.P.); (A.R.); (M.A.T.); (L.I.-S.); (M.M.)
| | - Marcio Alberto Torsoni
- Faculdade de Ciências Aplicadas, Universidade de Campinas, UNICAMP, Limeira 13484-350, São Paulo, Brazil; (M.d.N.B.); (L.A.d.P.S.); (J.P.C.); (C.P.); (A.R.); (M.A.T.); (L.I.-S.); (M.M.)
| | - Letícia Ignácio-Souza
- Faculdade de Ciências Aplicadas, Universidade de Campinas, UNICAMP, Limeira 13484-350, São Paulo, Brazil; (M.d.N.B.); (L.A.d.P.S.); (J.P.C.); (C.P.); (A.R.); (M.A.T.); (L.I.-S.); (M.M.)
| | - Marciane Milanski
- Faculdade de Ciências Aplicadas, Universidade de Campinas, UNICAMP, Limeira 13484-350, São Paulo, Brazil; (M.d.N.B.); (L.A.d.P.S.); (J.P.C.); (C.P.); (A.R.); (M.A.T.); (L.I.-S.); (M.M.)
| | - Michael G. Ross
- Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (M.G.R.); (M.D.)
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles at Harbor-UCLA, Torrance, CA 90502, USA
| | - Kelly Pereira Coca
- Ana Abrao Breastfeeding Center, Escola Paulista de Enfermagem, Universidade Federal São Paulo, UNIFESP, São Paulo 04037-001, São Paulo, Brazil;
| | - Mina Desai
- Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (M.G.R.); (M.D.)
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles at Harbor-UCLA, Torrance, CA 90502, USA
| | - Adriana Souza Torsoni
- Faculdade de Ciências Aplicadas, Universidade de Campinas, UNICAMP, Limeira 13484-350, São Paulo, Brazil; (M.d.N.B.); (L.A.d.P.S.); (J.P.C.); (C.P.); (A.R.); (M.A.T.); (L.I.-S.); (M.M.)
| |
Collapse
|
4
|
Kim OY, Song J. Important roles of linoleic acid and α-linolenic acid in regulating cognitive impairment and neuropsychiatric issues in metabolic-related dementia. Life Sci 2024; 337:122356. [PMID: 38123015 DOI: 10.1016/j.lfs.2023.122356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/02/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
Metabolic syndrome (MetS), which is characterized by insulin resistance, high blood glucose, obesity, and dyslipidemia, is known to increase the risk of dementia accompanied by memory loss and depression. The direct pathways and specific mechanisms in the central nervous system (CNS) for addressing fatty acid imbalances in MetS have not yet been fully elucidated. Among polyunsaturated acids, linoleic acid (LA, n6-PUFA) and α-linolenic acid (ALA, n3-PUFA), which are two essential fatty acids that should be provided by food sources (e.g., vegetable oils and seeds), have been reported to regulate various cellular mechanisms including apoptosis, inflammatory responses, mitochondrial biogenesis, and insulin signaling. Furthermore, inadequate intake of LA and ALA is reported to be involved in neuropathology and neuropsychiatric diseases as well as imbalanced metabolic conditions. Herein, we review the roles of LA and ALA on metabolic-related dementia focusing on insulin resistance, dyslipidemia, synaptic plasticity, cognitive function, and neuropsychiatric issues. This review suggests that LA and ALA are important fatty acids for concurrent treatment of both MetS and neurological problems.
Collapse
Affiliation(s)
- Oh Yoen Kim
- Department of Food Science and Nutrition, Dong A University, Busan, Republic of Korea; Department of Health Sciences, Graduate School of Dong-A University, Busan, Republic of Korea.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Zhou J, Zuo W, Tan Y, Wang X, Zhu M, Zhang H. Effects of n-3 polyunsaturated fatty acid on metabolic status in women with polycystic ovary syndrome: a meta-analysis of randomized controlled trials. J Ovarian Res 2023; 16:54. [PMID: 36932420 PMCID: PMC10022207 DOI: 10.1186/s13048-023-01130-4] [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: 05/12/2022] [Accepted: 02/27/2023] [Indexed: 03/19/2023] Open
Abstract
This meta-analysis was conducted to summarize the effects of n-3 polyunsaturated fatty acid (n-3 PUFA) on metabolic status including insulin metabolism and lipid metabolism in women with polycystic ovary syndrome (PCOS) by randomized controlled trials (RCTs). Four mainstream databases including PubMed, Cochrane Library, Embase and Web of Science were searched from their inception to October 2021. The registration number of this study was CRD42021285233. The quality assessment was performed referring the Cochrane Risk of Bias Tool. Mean differences (MD) and 95% confidence intervals (CIs) were generated for continuous variables by meta-analysis. Subgroup analyses were performed based on study duration (≤ 8 weeks or > 8 weeks), the source of n-3 PUFA (marine derived or plant origins) and dosage of n-3 PUFA (≤ 1000 mg/d or > 1000 mg/d). Eventually, 11 RCTs reporting 816 patients were enrolled. Compared with control group, n-3 PUFA treatment decreased waist circumference (MD = -2.76, 95% CI: -3.82 to -1.69; p < 0.00001), fasting plasma glucose (MD = -3.91, 95% CI: -5.69 to -2.13; p < 0.0001), fasting insulin (MD = -2.45, 95% CI: -3.19 to -1.71; p < 0.00001), homeostatic model assessment of insulin resistance (MD = -0.45, 95% CI: -0.80 to -0.11; p = 0.01), triglyceride (MD = -9.33, 95% CI: -10.56 to -8.10; p < 0.00001), total cholesterol (MD = -12.32, 95% CI: -19.15 to -5.50; p = 0.0004), low-density lipoprotein cholesterol (MD = -10.53, 95% CI: -19.31 to -1.75; p = 0.02), and increase quantitative insulin sensitivity check index (MD = 0.01, 95% CI: 0.01 to 0.02; p < 0.00001), Adiponectin (MD = 1.46, 95% CI: 1.12 to 1.80; p < 0.00001) in PCOS patients. However, n-3 PUFA failed to change body weight, body mass index, high-density lipoprotein cholesterol, very low-density lipoprotein cholesterol and hs-CRP in the overall analysis. Further subgroup analyses showed that supplements of n-3 PUFA for more than 8 weeks is more conducive to improve the metabolic status in insulin resistance and lipid profiles. The meta-analysis demonstrates that n-3 PUFA may be an effective intervention for alleviating metabolic status in PCOS. Hence, we recommend PCOS patients replenish n-3 PUFA with duration > 8 weeks regardless of the source and the dosage to retard the pathogenesis of PCOS related metabolic diseases.
Collapse
Grants
- project NO JC2020003 Project of Nantong Science and Technology Bureau, Jiangsu, China
- project NO JC2020003 Project of Nantong Science and Technology Bureau, Jiangsu, China
- project NO JC2020003 Project of Nantong Science and Technology Bureau, Jiangsu, China
- project NO JC2020003 Project of Nantong Science and Technology Bureau, Jiangsu, China
- project NO JC2020003 Project of Nantong Science and Technology Bureau, Jiangsu, China
- project NO JC2020003 Project of Nantong Science and Technology Bureau, Jiangsu, China
Collapse
Affiliation(s)
- Jie Zhou
- grid.410745.30000 0004 1765 1045Nanjing University of Chinese Medicine, Nanjing, 210023 People’s Republic of China
- Department of Chinese Medicine, The First People’s Hospital of Nantong, Nantong, 226001 Jiangsu People’s Republic of China
| | - Wenting Zuo
- grid.410745.30000 0004 1765 1045Department of Reproductive Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029 Jiangsu People’s Republic of China
| | - Yong Tan
- grid.410745.30000 0004 1765 1045Department of Reproductive Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029 Jiangsu People’s Republic of China
| | - Xudong Wang
- Department of Chinese Medicine, The First People’s Hospital of Nantong, Nantong, 226001 Jiangsu People’s Republic of China
| | - Meihong Zhu
- Department of Pharmacy, The First People’s Hospital of Nantong, Nantong, 226001 Jiangsu People’s Republic of China
| | - Huili Zhang
- Medical Department, The First People’s Hospital of Nantong, Nantong, 226001 Jiangsu People’s Republic of China
| |
Collapse
|
6
|
Specialized Pro-Resolving Mediators in Neuroinflammation: Overview of Studies and Perspectives of Clinical Applications. Molecules 2022; 27:molecules27154836. [PMID: 35956787 PMCID: PMC9370036 DOI: 10.3390/molecules27154836] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 11/24/2022] Open
Abstract
Specialized pro-resolving mediators (SPMs) are lipid mediators derived from poly-unsaturated fatty acids (PUFAs) which have been demonstrated to have an important role in the inflammation environment, preventing an overreaction of the organism and promoting the resolution of inflammation. Our purpose was to point out the current evidence for specialized pro-resolving mediators, focusing on their role in neuroinflammation and in major neurological diseases.
Collapse
|
7
|
Roles of Fatty Acids in Microglial Polarization: Evidence from In Vitro and In Vivo Studies on Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms23137300. [PMID: 35806302 PMCID: PMC9266841 DOI: 10.3390/ijms23137300] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
Microglial polarization to the M1 phenotype (classically activated) or the M2 phenotype (alternatively activated) is critical in determining the fate of immune responses in neurodegenerative diseases (NDs). M1 macrophages contribute to neurotoxicity, neuronal and synaptic damage, and oxidative stress and are the first line of defense, and M2 macrophages elicit an anti-inflammatory response to regulate neuroinflammation, clear cell debris, and promote neuroregeneration. Various studies have focused on the ability of natural compounds to promote microglial polarization from the M1 phenotype to the M2 phenotype in several diseases, including NDs. However, studies on the roles of fatty acids in microglial polarization and their implications in NDs are a rare find. Most of the studies support the role of polyunsaturated fatty acids (PUFAs) in microglial polarization using cell and animal models. Thus, we aimed to collect data and provide a narrative account of microglial types, markers, and studies pertaining to fatty acids, particularly PUFAs, on microglial polarization and their neuroprotective effects. The involvement of only PUFAs in the chosen topic necessitates more in-depth research into the role of unexplored fatty acids in microglial polarization and their mechanistic implications. The review also highlights limitations and future challenges.
Collapse
|
8
|
Dunn GA, Mitchell AJ, Selby M, Fair DA, Gustafsson HC, Sullivan EL. Maternal diet and obesity shape offspring central and peripheral inflammatory outcomes in juvenile non-human primates. Brain Behav Immun 2022; 102:224-236. [PMID: 35217175 PMCID: PMC8995380 DOI: 10.1016/j.bbi.2022.02.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 01/21/2022] [Accepted: 02/19/2022] [Indexed: 12/30/2022] Open
Abstract
The obesity epidemic affects 40% of adults in the US, with approximately one-third of pregnant women classified as obese. Previous research suggests that children born to obese mothers are at increased risk for a number of health conditions. The mechanisms behind this increased risk are poorly understood. Increased exposure to in-utero inflammation induced by maternal obesity is proposed as an underlying mechanism for neurodevelopmental alterations in offspring. Utilizing a non-human primate model of maternal obesity, we hypothesized that maternal consumption of an obesogenic diet will predict offspring peripheral (e.g., cytokines and chemokines) and central (microglia number) inflammatory outcomes via the diet's effects on maternal adiposity and maternal inflammatory state during the third trimester. We used structural equation modeling to simultaneously examine the complex associations among maternal diet, metabolic state, adiposity, inflammation, and offspring central and peripheral inflammation. Four latent variables were created to capture maternal chemokines and pro-inflammatory cytokines, and offspring cytokine and chemokines. Model results showed that offspring microglia counts in the basolateral amygdala were associated with maternal diet (β = -0.622, p < 0.01), adiposity (β = 0.593, p < 0.01), and length of gestation (β = 0.164, p < 0.05) but not with maternal chemokines (β = 0.135, p = 0.528) or maternal pro-inflammatory cytokines (β = 0.083, p = 0.683). Additionally, we found that juvenile offspring peripheral cytokines (β = -0.389, p < 0.01) and chemokines (β = -0.298, p < 0.05) were associated with a maternal adiposity-induced decrease in maternal circulating chemokines during the third trimester (β = -0.426, p < 0.01). In summary, these data suggest that maternal diet and adiposity appear to directly predict offspring amygdala microglial counts while maternal adiposity influences offspring peripheral inflammatory outcomes via maternal inflammatory state.
Collapse
Affiliation(s)
| | - A J Mitchell
- Oregon Health & Science University, Department of Behavioral Neuroscience, USA; Oregon National Primate Research Center, Department of Neuroscience, USA
| | - Matthew Selby
- University of Oregon, Department of Human Physiology, USA
| | - Damien A Fair
- University of Minnesota School of Medicine, Masonic Institute of Child Development, USA
| | | | - Elinor L Sullivan
- University of Oregon, Department of Human Physiology, USA; Oregon Health & Science University, Department of Behavioral Neuroscience, USA; Oregon National Primate Research Center, Department of Neuroscience, USA; Oregon Health & Science University, Department of Psychiatry, USA.
| |
Collapse
|
9
|
Mitchell AJ, Dunn GA, Sullivan EL. The Influence of Maternal Metabolic State and Nutrition on Offspring Neurobehavioral Development: A Focus on Preclinical Models. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:450-460. [PMID: 34915175 PMCID: PMC9086110 DOI: 10.1016/j.bpsc.2021.11.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/19/2021] [Accepted: 11/29/2021] [Indexed: 12/22/2022]
Abstract
The prevalence of both obesity and neurodevelopmental disorders has increased substantially over the last several decades. Early environmental factors, including maternal nutrition and metabolic state during gestation, influence offspring neurodevelopment. Both human and preclinical models demonstrate a link between poor maternal nutrition, altered metabolic state, and risk of behavioral abnormalities in offspring. This review aims to highlight evidence from the current literature connecting maternal nutrition and the associated metabolic changes with neural and behavioral outcomes in the offspring, as well as identify possible mechanisms underlying these neurodevelopmental outcomes. Owing to the highly correlated nature of poor nutrition and obesity in humans, preclinical animal models are important in distinguishing the unique effects of maternal nutrition and metabolic state on offspring brain development. We use a translational lens to highlight results from preclinical animal models of maternal obesogenic diet related to alterations in behavioral and neurodevelopmental outcomes in offspring. Specifically, we aim to highlight results that resemble behavioral phenotypes described in the diagnostic criteria of neurodevelopmental conditions in humans. Finally, we examine the proinflammatory nature of maternal obesity and consumption of a high-fat diet as a mechanism for neurodevelopmental alterations that may alter offspring behavior later in life. It is important that future studies examine potential therapeutic interventions and prevention strategies to interrupt the transgenerational transmission of the disease. Given the tremendous risk to the next generation, changes need to be made to ensure that all pregnant people have access to nutritious food and are informed about the optimal diet for their developing child.
Collapse
Affiliation(s)
- A J Mitchell
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon; Department of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon
| | - Geoffrey A Dunn
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Elinor L Sullivan
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon; Department of Psychiatry, Oregon Health & Science University, Portland, Oregon; Department of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon; Department of Human Physiology, University of Oregon, Eugene, Oregon.
| |
Collapse
|
10
|
Magalhaes MS, Potter HG, Ahlback A, Gentek R. Developmental programming of macrophages by early life adversity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 368:213-259. [PMID: 35636928 DOI: 10.1016/bs.ircmb.2022.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Macrophages are central elements of all organs, where they have a multitude of physiological and pathological functions. The first macrophages are produced during fetal development, and most adult organs retain populations of fetal-derived macrophages that self-maintain without major input of hematopoietic stem cell-derived monocytes. Their developmental origins make macrophages highly susceptible to environmental perturbations experienced in early life, in particular the fetal period. It is now well recognized that such adverse developmental conditions contribute to a wide range of diseases later in life. This chapter explores the notion that macrophages are key targets of environmental adversities during development, and mediators of their long-term impact on health and disease. We first briefly summarize our current understanding of macrophage ontogeny and their biology in tissues and consider potential mechanisms by which environmental stressors may mediate fetal programming. We then review evidence for programming of macrophages by adversities ranging from maternal immune activation and diet to environmental pollutants and toxins, which have disease relevance for different organ systems. Throughout this chapter, we contemplate appropriate experimental strategies to study macrophage programming. We conclude by discussing how our current knowledge of macrophage programming could be conceptualized, and finally highlight open questions in the field and approaches to address them.
Collapse
Affiliation(s)
- Marlene S Magalhaes
- Centre for Inflammation Research & Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Harry G Potter
- Centre for Inflammation Research & Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Anna Ahlback
- Centre for Inflammation Research & Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Rebecca Gentek
- Centre for Inflammation Research & Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
11
|
Susceptibility of Female Mice to the Dietary Omega-3/Omega-6 Fatty-Acid Ratio: Effects on Adult Hippocampal Neurogenesis and Glia. Int J Mol Sci 2022; 23:ijms23063399. [PMID: 35328825 PMCID: PMC8950413 DOI: 10.3390/ijms23063399] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 02/04/2023] Open
Abstract
Maternal intake of omega-3 (n-3 PUFAs) and omega-6 (n-6 PUFAs) polyunsaturated fatty acids impacts hippocampal neurogenesis during development, an effect that may extend to adulthood by altering adult hippocampal neurogenesis (AHN). The n-3 PUFAs and n-6 PUFAs are precursors of inflammatory regulators that potentially affect AHN and glia. Additionally, n-3 PUFA dietary supplementation may present a sexually dimorphic action in the brain. Therefore, we postulated that dietary n-6/n-3 PUFA balance shapes the adult DG in a sex-dependent manner influencing AHN and glia. We test our hypothesis by feeding adult female and male mice with n-3 PUFA balanced or deficient diets. To analyze the immunomodulatory potential of the diets, we injected mice with the bacterial endotoxin lipopolysaccharide (LPS). LPS reduced neuroblast number, and its effect was exacerbated by the n-3 PUFA-deficient diet. The n-3 PUFA-deficient diet reduced the DG volume, AHN, microglia number, and surveilled volume. The diet effect on most mature neuroblasts was exclusively significant in female mice. Colocalization and multivariate analysis revealed an association between microglia and AHN, as well as the sexual dimorphic effect of diet. Our study reveals that female mice are more susceptible than males to the effect of dietary n-6/n-3 PUFA ratio on AHN and microglia.
Collapse
|
12
|
Stress induced microglial activation contributes to depression. Pharmacol Res 2022; 179:106145. [DOI: 10.1016/j.phrs.2022.106145] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 02/08/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023]
|
13
|
Chumak T, Lecuyer MJ, Nilsson AK, Faustino J, Ardalan M, Svedin P, Sjöbom U, Ek J, Obenaus A, Vexler ZS, Mallard C. Maternal n-3 Polyunsaturated Fatty Acid Enriched Diet Commands Fatty Acid Composition in Postnatal Brain and Protects from Neonatal Arterial Focal Stroke. Transl Stroke Res 2021; 13:449-461. [PMID: 34674145 PMCID: PMC9046339 DOI: 10.1007/s12975-021-00947-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/19/2021] [Accepted: 09/13/2021] [Indexed: 01/20/2023]
Abstract
The fetus is strongly dependent on nutrients from the mother, including polyunsaturated fatty acids (PUFA). In adult animals, n-3 PUFA ameliorates stroke-mediated brain injury, but the modulatory effects of different PUFA content in maternal diet on focal arterial stroke in neonates are unknown. This study explored effects of maternal n-3 or n-6 enriched PUFA diets on neonatal stroke outcomes. Pregnant mice were assigned three isocaloric diets until offspring reached postnatal day (P) 10–13: standard, long-chain n-3 PUFA (n-3) or n-6 PUFA (n-6) enriched. Fatty acid profiles in plasma and brain of mothers and pups were determined by gas chromatography–mass spectrometry and cytokines/chemokines by multiplex protein analysis. Transient middle cerebral artery occlusion (tMCAO) was induced in P9-10 pups and cytokine and chemokine accumulation, caspase-3 and calpain-dependent spectrin cleavage and brain infarct volume were analyzed. The n-3 diet uniquely altered brain lipid profile in naïve pups. In contrast, cytokine and chemokine levels did not differ between n-3 and n-6 diet in naïve pups. tMCAO triggered accumulation of inflammatory cytokines and caspase-3-dependent and -independent cell death in ischemic-reperfused regions in pups regardless of diet, but magnitude of neuroinflammation and caspase-3 activation were attenuated in pups on n-3 diet, leading to protection against neonatal stroke. In conclusion, maternal/postnatal n-3 enriched diet markedly rearranges neonatal brain lipid composition and modulates the response to ischemia. While standard diet is sufficient to maintain low levels of inflammatory cytokines and chemokines under physiological conditions, n-3 PUFA enriched diet, but not standard diet, attenuates increases of inflammatory cytokines and chemokines in ischemic-reperfused regions and protects from neonatal stroke.
Collapse
Affiliation(s)
- Tetyana Chumak
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | | | - Anders K Nilsson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joel Faustino
- Department of Neurology, UCSF, San Francisco, CA, USA
| | - Maryam Ardalan
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Pernilla Svedin
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Ulrika Sjöbom
- Institute of Health and Care Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joakim Ek
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Andre Obenaus
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| | | | - Carina Mallard
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden. .,Department of Pediatrics, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
14
|
Wang CC, Du L, Shi HH, Ding L, Yanagita T, Xue CH, Wang YM, Zhang TT. Dietary EPA-Enriched Phospholipids Alleviate Chronic Stress and LPS-Induced Depression- and Anxiety-Like Behavior by Regulating Immunity and Neuroinflammation. Mol Nutr Food Res 2021; 65:e2100009. [PMID: 34219360 DOI: 10.1002/mnfr.202100009] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 06/10/2021] [Indexed: 12/17/2022]
Abstract
SCOPE A growing number of studies have reported the effects of eicosapentaenoic acid (EPA) and terrestrial phospholipids on ameliorating mood disorders. Marine-derived EPA-enriched phospholipids (EPA-PL) exhibit the structural characteristics of EPA and phospholipids. However, the effect of dietary EPA-PL, and the differences between amphiphilic EPA-PL and lyophobic EPA on mood disorders had not been studied. METHODS AND RESULTS A comparative investigation to determine the effects of dietary EPA-enriched ethyl ester (EPA-EE) and EPA-PL on improving depression- and anxiety-like behavior in a mouse model is performed, induced by 4 week chronic unpredictable mild stress (CUMS) coupled with lipopolysaccharide (LPS) challenge. It is found that dietary 4 week 0.6% (w/w) EPA-PL rescued depression- and anxiety-like behavior to a greater extent than did EPA-EE. Moreover, dietary EPA-PL significantly reduced the immobility time by 56.6%, close to the normal level, in forced swimming test, which revealed a reversal of depression-like behavior. Further studies revealed that dietary EPA-PL regulated immunity, monoamine systems, and the hypothalamic-pituitary-adrenal (HPA) axis by multi-target interactions, including inhibition of neuroinflammation and apoptosis. CONCLUSION EPA-PL exerted superior effects to EPA-EE in alleviating depression- and anxiety-like behavior. The data suggest potential novel candidate or targeted dietary patterns to prevent and treat mood disorder.
Collapse
Affiliation(s)
- Cheng-Cheng Wang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, P. R. China
| | - Lei Du
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, No. 44 Wenhuaxi Road, Jinan, Shandong, 250012, P. R. China
| | - Hao-Hao Shi
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, P. R. China
| | - Lin Ding
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, P. R. China
| | - Teruyoshi Yanagita
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong Province, 266237, P. R. China
| | - Chang-Hu Xue
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, P. R. China
- Laboratory of Nutrition Biochemistry, Department of Applied Biochemistry and Food Science, Saga University, Saga, 840-8502, Japan
| | - Yu-Ming Wang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, P. R. China
- Laboratory of Nutrition Biochemistry, Department of Applied Biochemistry and Food Science, Saga University, Saga, 840-8502, Japan
| | - Tian-Tian Zhang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, P. R. China
| |
Collapse
|
15
|
Murine and human microglial cells are relatively enriched with eicosapentaenoic acid compared to the whole brain. Neurochem Int 2021; 150:105154. [PMID: 34384851 DOI: 10.1016/j.neuint.2021.105154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/04/2021] [Accepted: 08/07/2021] [Indexed: 12/30/2022]
Abstract
The brain is a multicellular organ enriched with lipids. While the fatty acid composition of gross cerebral tissue is well characterized, the fatty acid composition of specific brain cells, particularly microglia cells, is less well characterized. Microglia cells are the innate immune cells of the brain, and a paucity of studies measuring their fatty acid composition using either immortalized or primary microglia cells report a higher ratio of eicosapentaenoic acid (EPA) to docosahexaenoic acid (DHA) than widely observed in whole brain tissue. Here we further characterize the fatty acid composition of murine microglia cells from young male and female mice as well as of human origin and compared it with a myelin-enriched fraction from the same mice. Our results show that saturated and monounsaturated fatty acids are the most abundant followed by polyunsaturated fatty acids (PUFA), with no statistical differences between sexes. Regarding PUFA, although DHA levels did not differ between human and murine cells, EPA was statistically higher in murine microglia. Notably, the DHA to EPA ratio was about 400 times higher in microglial cells compared to the myelin-enriched fraction. Thus, our results suggest that as compared to whole brain tissue EPA is relatively abundant in microglia cells, particularly in comparison to other n-3 PUFA such as DHA. Since the fatty acid composition of microglia can influence their functionality, a better understanding of EPA and DHA metabolism in microglia and the brain could identify new targets to modify microglial activity.
Collapse
|
16
|
Disentangling the Molecular Mechanisms of the Antidepressant Activity of Omega-3 Polyunsaturated Fatty Acid: A Comprehensive Review of the Literature. Int J Mol Sci 2021; 22:ijms22094393. [PMID: 33922396 PMCID: PMC8122828 DOI: 10.3390/ijms22094393] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/20/2021] [Accepted: 04/20/2021] [Indexed: 12/12/2022] Open
Abstract
Major depressive disorders (MDDs) are often associated with a deficiency in long-chain omega-3 polyunsaturated fatty acids (ω-3 PUFAs), as well as signs of low-grade inflammation. Epidemiological and dietary studies suggest that a high intake of fish, the major source of ω-3 PUFAs, is associated with lower rates of MDDs. Meta-analyses of randomized placebo-controlled ω-3 PUFAs intervention-trials suggest that primarily eicosapentaenoic acid (EPA), but not docosahexaenoic acid (DHA), is responsible for the proposed antidepressant effect. In this review, we dissect the current biological knowledge on EPA and DHA and their bioactive lipid metabolites to search for a pharmacological explanation of this, to date, unexplained clinical observation. Through enzymatic conversion by cyclooxygenase (COX), lipoxygenase (ALOX), and cytochrome P-450 monooxygenase (CYP), EPA and DHA are metabolized to major anti-inflammatory and pro-resolving lipid mediators. In addition, both ω-3 PUFAs are precursors for endocannabinoids, with known effects on immunomodulation, neuroinflammation, food intake and mood. Finally, both ω-3 PUFAs are crucial for the structure and organization of membranes and lipid rafts. While most biological effects are shared by these two ω-3 PUFAs, some distinct features could be identified: (1) The preferential CYP monooxygenase pathway for EPA and EPA derived eicosanoids; (2) The high CB2 receptor affinities of EPA-derived EPEA and its epoxy-metabolite 17,18-EEQ-EA, while the DHA-derived endocannabinoids lack such receptor affinities; (3) The competition of EPA but not DHA with arachidonic acid (AA) for particular glycerophospholipids. EPA and AA are preferentially incorporated into phosphatidylinositols, while DHA is mainly incorporated into phosphatidyl-ethanolamine, -serine and -choline. We propose that these distinct features may explain the superior antidepressant activity of EPA rich ω-3 PUFAs and that these are potential novel targets for future antidepressant drugs.
Collapse
|
17
|
Davis J, Mire E. Maternal obesity and developmental programming of neuropsychiatric disorders: An inflammatory hypothesis. Brain Neurosci Adv 2021; 5:23982128211003484. [PMID: 33889757 PMCID: PMC8040564 DOI: 10.1177/23982128211003484] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 12/19/2022] Open
Abstract
Maternal obesity is associated with the development of a variety of neuropsychiatric disorders; however, the mechanisms behind this association are not fully understood. Comparison between maternal immune activation and maternal obesity reveals similarities in associated impairments and maternal cytokine profile. Here, we present a summary of recent evidence describing how inflammatory processes contribute towards the development of neuropsychiatric disorders in the offspring of obese mothers. This includes discussion on how maternal cytokine levels, fatty acids and placental inflammation may interact with foetal neurodevelopment through changes to microglial behaviour and epigenetic modification. We also propose an exosome-mediated mechanism for the disruption of brain development under maternal obesity and discuss potential intervention strategies.
Collapse
Affiliation(s)
- Jonathan Davis
- Hodge Centre for Neuropsychiatric Immunology, Neuroscience and Mental Health Research Institute, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Erik Mire
- Hodge Centre for Neuropsychiatric Immunology, Neuroscience and Mental Health Research Institute, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
18
|
Martinat M, Rossitto M, Di Miceli M, Layé S. Perinatal Dietary Polyunsaturated Fatty Acids in Brain Development, Role in Neurodevelopmental Disorders. Nutrients 2021; 13:1185. [PMID: 33918517 PMCID: PMC8065891 DOI: 10.3390/nu13041185] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 12/26/2022] Open
Abstract
n-3 and n-6 polyunsaturated fatty acids (PUFAs) are essential fatty acids that are provided by dietary intake. Growing evidence suggests that n-3 and n-6 PUFAs are paramount for brain functions. They constitute crucial elements of cellular membranes, especially in the brain. They are the precursors of several metabolites with different effects on inflammation and neuron outgrowth. Overall, long-chain PUFAs accumulate in the offspring brain during the embryonic and post-natal periods. In this review, we discuss how they accumulate in the developing brain, considering the maternal dietary supply, the polymorphisms of genes involved in their metabolism, and the differences linked to gender. We also report the mechanisms linking their bioavailability in the developing brain, their transfer from the mother to the embryo through the placenta, and their role in brain development. In addition, data on the potential role of altered bioavailability of long-chain n-3 PUFAs in the etiologies of neurodevelopmental diseases, such as autism, attention deficit and hyperactivity disorder, and schizophrenia, are reviewed.
Collapse
|
19
|
Leyrolle Q, Decoeur F, Briere G, Amadieu C, Quadros ARAA, Voytyuk I, Lacabanne C, Benmamar-Badel A, Bourel J, Aubert A, Sere A, Chain F, Schwendimann L, Matrot B, Bourgeois T, Grégoire S, Leblanc JG, De Moreno De Leblanc A, Langella P, Fernandes GR, Bretillon L, Joffre C, Uricaru R, Thebault P, Gressens P, Chatel JM, Layé S, Nadjar A. Maternal dietary omega-3 deficiency worsens the deleterious effects of prenatal inflammation on the gut-brain axis in the offspring across lifetime. Neuropsychopharmacology 2021; 46:579-602. [PMID: 32781459 PMCID: PMC8026603 DOI: 10.1038/s41386-020-00793-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/16/2020] [Accepted: 07/27/2020] [Indexed: 12/18/2022]
Abstract
Maternal immune activation (MIA) and poor maternal nutritional habits are risk factors for the occurrence of neurodevelopmental disorders (NDD). Human studies show the deleterious impact of prenatal inflammation and low n-3 polyunsaturated fatty acid (PUFA) intake on neurodevelopment with long-lasting consequences on behavior. However, the mechanisms linking maternal nutritional status to MIA are still unclear, despite their relevance to the etiology of NDD. We demonstrate here that low maternal n-3 PUFA intake worsens MIA-induced early gut dysfunction, including modification of gut microbiota composition and higher local inflammatory reactivity. These deficits correlate with alterations of microglia-neuron crosstalk pathways and have long-lasting effects, both at transcriptional and behavioral levels. This work highlights the perinatal period as a critical time window, especially regarding the role of the gut-brain axis in neurodevelopment, elucidating the link between MIA, poor nutritional habits, and NDD.
Collapse
Affiliation(s)
- Q. Leyrolle
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France ,Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France
| | - F. Decoeur
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - G. Briere
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France ,grid.503269.b0000 0001 2289 8198CNRS, Bordeaux INP, LaBRI, UMR 5800, F-33400 Talence, France
| | - C. Amadieu
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. R. A. A. Quadros
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - I. Voytyuk
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - C. Lacabanne
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. Benmamar-Badel
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - J. Bourel
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. Aubert
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. Sere
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - F. Chain
- grid.460789.40000 0004 4910 6535Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - L. Schwendimann
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France
| | - B. Matrot
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France
| | - T. Bourgeois
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France
| | - S. Grégoire
- grid.462804.c0000 0004 0387 2525Centre des Sciences du Goût et de l’Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France
| | - J. G. Leblanc
- CERELA-CONICET, San Miguel de Tucuman, 4000 Tucuman, Argentina
| | | | - P. Langella
- grid.460789.40000 0004 4910 6535Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - G. R. Fernandes
- Rene Rachou Institute – Oswaldo Cruz Foundation, Belo Horizonte, MG Brazil
| | - L. Bretillon
- grid.462804.c0000 0004 0387 2525Centre des Sciences du Goût et de l’Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France
| | - C. Joffre
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - R. Uricaru
- grid.503269.b0000 0001 2289 8198CNRS, Bordeaux INP, LaBRI, UMR 5800, F-33400 Talence, France
| | - P. Thebault
- grid.503269.b0000 0001 2289 8198CNRS, Bordeaux INP, LaBRI, UMR 5800, F-33400 Talence, France
| | - P. Gressens
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France ,grid.13097.3c0000 0001 2322 6764Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King’s College London, King’s Health Partners, St. Thomas’ Hospital, London, SE1 7EH UK
| | - J. M. Chatel
- grid.460789.40000 0004 4910 6535Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - S. Layé
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. Nadjar
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| |
Collapse
|
20
|
Bordeleau M, Fernández de Cossío L, Chakravarty MM, Tremblay MÈ. From Maternal Diet to Neurodevelopmental Disorders: A Story of Neuroinflammation. Front Cell Neurosci 2021; 14:612705. [PMID: 33536875 PMCID: PMC7849357 DOI: 10.3389/fncel.2020.612705] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Providing the appropriate quantity and quality of food needed for both the mother's well-being and the healthy development of the offspring is crucial during pregnancy. However, the macro- and micronutrient intake also impacts the body's regulatory supersystems of the mother, such as the immune, endocrine, and nervous systems, which ultimately influence the overall development of the offspring. Of particular importance is the association between unhealthy maternal diet and neurodevelopmental disorders in the offspring. Epidemiological studies have linked neurodevelopmental disorders like autism spectrum disorders, attention-deficit-hyperactivity disorder, and schizophrenia, to maternal immune activation (MIA) during gestation. While the deleterious consequences of diet-induced MIA on offspring neurodevelopment are increasingly revealed, neuroinflammation is emerging as a key underlying mechanism. In this review, we compile the evidence available on how the mother and offspring are both impacted by maternal dietary imbalance. We specifically explore the various inflammatory and anti-inflammatory effects of dietary components and discuss how changes in inflammatory status can prime the offspring brain development toward neurodevelopmental disorders. Lastly, we discuss research evidence on the mechanisms that sustain the relationship between maternal dietary imbalance and offspring brain development, involving altered neuroinflammatory status in the offspring, as well as genetic to cellular programming notably of microglia, and the evidence that the gut microbiome may act as a key mediator.
Collapse
Affiliation(s)
- Maude Bordeleau
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | | | - M. Mallar Chakravarty
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Cerebral Imaging Centre, Douglas Mental Health University, McGill University, Montréal, QC, Canada
- Department of Psychiatry, McGill University, Montréal, QC, Canada
- Department of Biological and Biomedical Engineering, McGill University, Montréal, QC, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Biochemistry and Molecular Biology, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
21
|
Madore C, Leyrolle Q, Morel L, Rossitto M, Greenhalgh AD, Delpech JC, Martinat M, Bosch-Bouju C, Bourel J, Rani B, Lacabanne C, Thomazeau A, Hopperton KE, Beccari S, Sere A, Aubert A, De Smedt-Peyrusse V, Lecours C, Bisht K, Fourgeaud L, Gregoire S, Bretillon L, Acar N, Grant NJ, Badaut J, Gressens P, Sierra A, Butovsky O, Tremblay ME, Bazinet RP, Joffre C, Nadjar A, Layé S. Essential omega-3 fatty acids tune microglial phagocytosis of synaptic elements in the mouse developing brain. Nat Commun 2020; 11:6133. [PMID: 33257673 PMCID: PMC7704669 DOI: 10.1038/s41467-020-19861-z] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/03/2020] [Indexed: 12/23/2022] Open
Abstract
Omega-3 fatty acids (n-3 PUFAs) are essential for the functional maturation of the brain. Westernization of dietary habits in both developed and developing countries is accompanied by a progressive reduction in dietary intake of n-3 PUFAs. Low maternal intake of n-3 PUFAs has been linked to neurodevelopmental diseases in Humans. However, the n-3 PUFAs deficiency-mediated mechanisms affecting the development of the central nervous system are poorly understood. Active microglial engulfment of synapses regulates brain development. Impaired synaptic pruning is associated with several neurodevelopmental disorders. Here, we identify a molecular mechanism for detrimental effects of low maternal n-3 PUFA intake on hippocampal development in mice. Our results show that maternal dietary n-3 PUFA deficiency increases microglia-mediated phagocytosis of synaptic elements in the rodent developing hippocampus, partly through the activation of 12/15-lipoxygenase (LOX)/12-HETE signaling, altering neuronal morphology and affecting cognitive performance of the offspring. These findings provide a mechanistic insight into neurodevelopmental defects caused by maternal n-3 PUFAs dietary deficiency.
Collapse
Affiliation(s)
- C Madore
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women´s Hospital, Harvard Medical School, Boston, MA, USA
| | - Q Leyrolle
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
- NeuroDiderot, Inserm, Université de Paris Diderot, F-75019, Paris, France
| | - L Morel
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - M Rossitto
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - A D Greenhalgh
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - J C Delpech
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - M Martinat
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - C Bosch-Bouju
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - J Bourel
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - B Rani
- Department of Health Sciences, University of Florence, Florence, Italy
| | - C Lacabanne
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - A Thomazeau
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - K E Hopperton
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, M5S 3E2, Canada
| | - S Beccari
- Achucarro Basque Center for Neuroscience, University of the Basque Country and Ikerbasque Foundation, 48940, Leioa, Spain
| | - A Sere
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - A Aubert
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - V De Smedt-Peyrusse
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - C Lecours
- Neurosciences Axis, CRCHU de Québec-Université Laval, Québec City, QC, Canada
| | - K Bisht
- Neurosciences Axis, CRCHU de Québec-Université Laval, Québec City, QC, Canada
| | - L Fourgeaud
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - S Gregoire
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Univ. Bourgogne Franche-Comté, F-21000, Dijon, France
| | - L Bretillon
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Univ. Bourgogne Franche-Comté, F-21000, Dijon, France
| | - N Acar
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Univ. Bourgogne Franche-Comté, F-21000, Dijon, France
| | - N J Grant
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - J Badaut
- CNRS UMR5287, University of Bordeaux, Bordeaux, France
| | - P Gressens
- NeuroDiderot, Inserm, Université de Paris Diderot, F-75019, Paris, France
- Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, SE1 7EH, UK
| | - A Sierra
- Achucarro Basque Center for Neuroscience, University of the Basque Country and Ikerbasque Foundation, 48940, Leioa, Spain
| | - O Butovsky
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women´s Hospital, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - M E Tremblay
- Neurosciences Axis, CRCHU de Québec-Université Laval, Québec City, QC, Canada
| | - R P Bazinet
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, M5S 3E2, Canada
| | - C Joffre
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - A Nadjar
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France.
| | - S Layé
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France.
| |
Collapse
|
22
|
Di Miceli M, Bosch-Bouju C, Layé S. PUFA and their derivatives in neurotransmission and synapses: a new hallmark of synaptopathies. Proc Nutr Soc 2020; 79:1-16. [PMID: 32299516 DOI: 10.1017/s0029665120000129] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PUFA of the n-3 and n-6 families are present in high concentration in the brain where they are major components of cell membranes. The main forms found in the brain are DHA (22 :6, n-3) and arachidonic acid (20:4, n-6). In the past century, several studies pinpointed that modifications of n-3 and n-6 PUFA levels in the brain through dietary supply or genetic means are linked to the alterations of synaptic function. Yet, synaptopathies emerge as a common characteristic of neurodevelopmental disorders, neuropsychiatric diseases and some neurodegenerative diseases. Understanding the mechanisms of action underlying the activity of PUFA at the level of synapses is thus of high interest. In this frame, dietary supplementation in PUFA aiming at restoring or promoting the optimal function of synapses appears as a promising strategy to treat synaptopathies. This paper reviews the link between dietary PUFA, synapse formation and the role of PUFA and their metabolites in synaptic functions.
Collapse
Affiliation(s)
- Mathieu Di Miceli
- INRAE, University of Bordeaux, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - Clémentine Bosch-Bouju
- INRAE, University of Bordeaux, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - Sophie Layé
- INRAE, University of Bordeaux, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| |
Collapse
|
23
|
Bazinet RP, Metherel AH, Chen CT, Shaikh SR, Nadjar A, Joffre C, Layé S. Brain eicosapentaenoic acid metabolism as a lead for novel therapeutics in major depression. Brain Behav Immun 2020; 85:21-28. [PMID: 31278982 DOI: 10.1016/j.bbi.2019.07.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/26/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022] Open
Abstract
The results of several meta-analyses suggest that eicosapentaenoic acid (EPA) supplementation is therapeutic in managing the symptoms of major depression. It was previously assumed that because EPA is extremely low in the brain it did not cross the blood-brain barrier and any therapeutic effects it exerted would be via the periphery. However, more recent studies have established that EPA does enter the brain, but is rapidly metabolised following entry. While EPA does not accumulate within the brain, it is present in microglia and homeostatic mechanisms may regulate its esterification to phospholipids that serve important roles in cell signaling. Furthermore, a variety of signaling molecules from EPA have been described in the periphery and they have the potential to exert effects within the brain. If EPA is confirmed to be therapeutic in major depression as a result of adequately powered randomized clinical trials, future research on brain EPA metabolism could lead to the discovery of novel targets for treating or preventing major depression.
Collapse
Affiliation(s)
- Richard P Bazinet
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Canada.
| | - Adam H Metherel
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Chuck T Chen
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, North Bethesda, MD 20852, United States
| | - Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public Health & School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Agnes Nadjar
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Université de Bordeaux 2, 146 rue Léo Saignat, 33076 Bordeaux Cedex, France
| | - Corinne Joffre
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Université de Bordeaux 2, 146 rue Léo Saignat, 33076 Bordeaux Cedex, France
| | - Sophie Layé
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Université de Bordeaux 2, 146 rue Léo Saignat, 33076 Bordeaux Cedex, France
| |
Collapse
|
24
|
Effects of Docosahexaenoic Acid and Its Peroxidation Product on Amyloid-β Peptide-Stimulated Microglia. Mol Neurobiol 2019; 57:1085-1098. [PMID: 31677009 DOI: 10.1007/s12035-019-01805-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 09/26/2019] [Indexed: 12/13/2022]
Abstract
Growing evidence suggests that docosahexaenoic acid (DHA) exerts neuroprotective effects, although the mechanism(s) underlying these beneficial effects are not fully understood. Here we demonstrate that DHA, but not arachidonic acid (ARA), suppressed oligomeric amyloid-β peptide (oAβ)-induced reactive oxygen species (ROS) production in primary mouse microglia and immortalized mouse microglia (BV2). Similarly, DHA but not ARA suppressed oAβ-induced increases in phosphorylated cytosolic phospholipase A2 (p-cPLA2), inducible nitric oxide synthase (iNOS), and tumor necrosis factor-α (TNF-α) in BV2 cells. LC-MS/MS assay indicated the ability for DHA to cause an increase in 4-hydroxyhexenal (4-HHE) and suppress oAβ-induced increase in 4-hydroxynonenal (4-HNE). Although oAβ did not alter the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, exogenous DHA, ARA as well as low concentrations of 4-HHE and 4-HNE upregulated this pathway and increased production of heme oxygenase-1 (HO-1) in microglial cells. These results suggest that DHA modulates ARA metabolism in oAβ-stimulated microglia through suppressing oxidative and inflammatory pathways and upregulating the antioxidative stress pathway involving Nrf2/HO-1. Understanding the mechanism(s) underlying the beneficial effects of DHA on microglia should shed light into nutraceutical therapy for the prevention and treatment of Alzheimer's disease (AD).
Collapse
|
25
|
Safi-Stibler S, Gabory A. Epigenetics and the Developmental Origins of Health and Disease: Parental environment signalling to the epigenome, critical time windows and sculpting the adult phenotype. Semin Cell Dev Biol 2019; 97:172-180. [PMID: 31587964 DOI: 10.1016/j.semcdb.2019.09.008] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 09/19/2019] [Accepted: 09/26/2019] [Indexed: 02/06/2023]
Abstract
The literature about Developmental Origins of Health and Disease (DOHaD) studies is considerably growing. Maternal and paternal environment, during all the development of the individual from gametogenesis to weaning and beyond, as well as the psychosocial environment in childhood and teenage, can shape the adult and the elderly person's susceptibility to her/his own environment and diseases. This non-conventional, non-genetic, inheritance is underlain by several mechanisms among which epigenetics is obviously central, due to the notion of memory of early decisional events during development even when this stimulus is gone, that is implied in Waddington's developmental concept. This review first summarizes the different mechanisms by which the environment can model the epigenome: receptor signalling, energy metabolism and signal mechanotransduction from extracellular matrix to chromatin. Then an overview of the epigenetic changes in response to maternal environment during the vulnerability time windows, gametogenesis, early development, placentation and foetal growth, and postnatal period, is described, with the specific example of overnutrition and food deprivation. The implication of epigenetics in DOHaD is obvious, however the precise causal chain from early environment to the epigenome modifications to the phenotype still needs to be deciphered.
Collapse
Affiliation(s)
- Sofiane Safi-Stibler
- UMR BDR, INRA, ENVA, Université Paris Saclay, 78350, Jouy-en-Josas, France; Sorbonne Université, Collège Doctoral, F-75005, Paris, France
| | - Anne Gabory
- UMR BDR, INRA, ENVA, Université Paris Saclay, 78350, Jouy-en-Josas, France.
| |
Collapse
|
26
|
Caputo MP, Radlowski EC, Lawson M, Antonson A, Watson JE, Matt SM, Leyshon BJ, Das A, Johnson RW. Herring roe oil supplementation alters microglial cell gene expression and reduces peripheral inflammation after immune activation in a neonatal piglet model. Brain Behav Immun 2019; 81:455-469. [PMID: 31271868 PMCID: PMC6754775 DOI: 10.1016/j.bbi.2019.06.046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/20/2019] [Accepted: 06/29/2019] [Indexed: 01/29/2023] Open
Abstract
Neonatal brain development can be disrupted by infection that results in microglial cell activation and neuroinflammation. Studies indicate that polyunsaturated fatty acids (PUFAs) and their metabolites can resolve inflammation. It is not known if dietary PUFA increases lipid metabolites in brain or reduces neuroinflammation in neonates. We hypothesized that dietary PUFAs might suppress neuroinflammation by inhibiting pro-inflammatory cytokine over-production and promoting inflammatory resolution in the periphery and brain. Piglets were obtained on postnatal day (PD) 2 and randomly assigned to herring roe oil (HRO) or control (CON) diet. HRO was included at 2 g/kg powdered diet. HRO increased DHA levels in occipital lobe and the DHA to arachidonic acid (ARA) ratio in hippocampal tissue. HRO decreased ARA metabolites in occipital lobe. HRO failed to attenuate microglial pro-inflammatory cytokine production ex vivo. HRO did not affect fever or circulating resolvin D1 levels. HRO decreased circulating neutrophils and liver inflammatory gene expression, but increased resolution marker gene expression in liver post LPS. HRO upregulated CXCL16, TGFBR1, and C1QA in microglial cells. HRO supplementation exerted beneficial effects on inflammation in the periphery, but further studies are needed to evaluate the specific effects of omega-3 supplementation on microglial cell physiology in the neonate.
Collapse
Affiliation(s)
- Megan P. Caputo
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 449 Bevier Hall, 905 South Goodwin Ave, Urbana, IL, 61802 USA,Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 West Gregory Dr., Urbana, IL, 61802 USA,Veterinary Medical Scholars Program, Office of Research and Advanced Studies, University of Illinois at Urbana-Champaign, College of Veterinary Medicine, 3505 VMBSB, 2001 South Lincoln Ave, Urbana, IL, 61802 USA
| | - Emily C. Radlowski
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 449 Bevier Hall, 905 South Goodwin Ave, Urbana, IL, 61802 USA,Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 West Gregory Dr., Urbana, IL, 61802 USA
| | - Marcus Lawson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 West Gregory Dr., Urbana, IL, 61802 USA
| | - Adrienne Antonson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 West Gregory Dr., Urbana, IL, 61802 USA
| | - Josephine E. Watson
- Department of Biochemistry, School of Molecular & Cellular Biology, University of Illinois at Urbana-Champaign, 393 Morrill Hall, 505 South Goodwin Ave, Urbana, IL, 61802 USA
| | - Stephanie M. Matt
- Neuroscience Program, University of Illinois at Urbana-Champaign, 2325/21 Beckman Institute, 405 North Matthews Ave, Urbana, IL, 61801 USA
| | - Brian J. Leyshon
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 449 Bevier Hall, 905 South Goodwin Ave, Urbana, IL, 61802 USA,Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 West Gregory Dr., Urbana, IL, 61802 USA
| | - Aditi Das
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 449 Bevier Hall, 905 South Goodwin Ave, Urbana, IL 61802, USA; Department of Biochemistry, School of Molecular & Cellular Biology, University of Illinois at Urbana-Champaign, 393 Morrill Hall, 505 South Goodwin Ave, Urbana, IL 61802, USA; Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, 3516 VMBSB, 2001 South Lincoln Ave, Urbana, IL 61802, USA; Neuroscience Program, University of Illinois at Urbana-Champaign, 2325/21 Beckman Institute, 405 North Matthews Ave, Urbana, IL 61801, USA; Bioengineering Department, University of Illinois at Urbana-Champaign, 1102 Everitt Lab, MC-278, 1406 West Green St., Urbana, IL 61801, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 North Matthews Ave, M/C 251, Urbana, IL 61801, USA.
| | - Rodney W. Johnson
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 449 Bevier Hall, 905 South Goodwin Ave, Urbana, IL, 61802 USA,Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 West Gregory Dr., Urbana, IL, 61802 USA,Neuroscience Program, University of Illinois at Urbana-Champaign, 2325/21 Beckman Institute, 405 North Matthews Ave, Urbana, IL, 61801 USA
| |
Collapse
|
27
|
Joffre C, Rey C, Layé S. N-3 Polyunsaturated Fatty Acids and the Resolution of Neuroinflammation. Front Pharmacol 2019; 10:1022. [PMID: 31607902 PMCID: PMC6755339 DOI: 10.3389/fphar.2019.01022] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 08/12/2019] [Indexed: 12/22/2022] Open
Abstract
In the past few decades, as a result of their anti-inflammatory properties, n-3 long chain polyunsaturated fatty acids (n-3 LC-PUFAs), have gained greater importance in the regulation of inflammation, especially in the central nervous system (in this case known as neuroinflammation). If sustained, neuroinflammation is a common denominator of neurological disorders, including Alzheimer’s disease and major depression, and of aging. Hence, limiting neuroinflammation is a real strategy for neuroinflammatory disease therapy and treatment. Recent data show that n-3 LC-PUFAs exert anti-inflammatory properties in part through the synthesis of specialized pro-resolving mediators (SPMs) such as resolvins, maresins and protectins. These SPMs are crucially involved in the resolution of inflammation. They could be good candidates to resolve brain inflammation and to contribute to neuroprotective functions and could lead to novel therapeutics for brain inflammatory diseases. This review presents an overview 1) of brain n-3 LC-PUFAs as precursors of SPMs with an emphasis on the effect of n-3 PUFAs on neuroinflammation, 2) of the formation and action of SPMs in the brain and their biological roles, and the possible regulation of their synthesis by environmental factors such as inflammation and nutrition and, in particular, PUFA consumption.
Collapse
Affiliation(s)
- Corinne Joffre
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France.,Université de Bordeaux 2, Bordeaux, France
| | - Charlotte Rey
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France.,Université de Bordeaux 2, Bordeaux, France.,ITERG, Nutrition Health and Lipid Biochemistry Department, Canéjan, France
| | - Sophie Layé
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France.,Université de Bordeaux 2, Bordeaux, France
| |
Collapse
|
28
|
Bobbo VCD, Jara CP, Mendes NF, Morari J, Velloso LA, Araújo EP. Interleukin-6 Expression by Hypothalamic Microglia in Multiple Inflammatory Contexts: A Systematic Review. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1365210. [PMID: 31534953 PMCID: PMC6724433 DOI: 10.1155/2019/1365210] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/25/2019] [Indexed: 12/14/2022]
Abstract
Interleukin-6 (IL-6) is a unique cytokine that can play both pro- and anti-inflammatory roles depending on the anatomical site and conditions under which it has been induced. Specific neurons of the hypothalamus provide important signals to control food intake and energy expenditure. In individuals with obesity, a microglia-dependent inflammatory response damages the neural circuits responsible for maintaining whole-body energy homeostasis, resulting in a positive energy balance. However, little is known about the role of IL-6 in the regulation of hypothalamic microglia. In this systematic review, we asked what types of conditions and stimuli could modulate microglial IL-6 expression in murine model. We searched the PubMed and Web of Science databases and analyzed 13 articles that evaluated diverse contexts and study models focused on IL-6 expression and microglia activation, including the effects of stress, hypoxia, infection, neonatal overfeeding and nicotine exposure, lipopolysaccharide stimulus, hormones, exercise protocols, and aging. The results presented in this review emphasized the role of "injury-like" stimuli, under which IL-6 acts as a proinflammatory cytokine, concomitant with marked microglial activation, which drive hypothalamic neuroinflammation. Emerging evidence indicates an important correlation of basal IL-6 levels and microglial function with the maintenance of hypothalamic homeostasis. Advances in our understanding of these different contexts will lead to the development of more specific pharmacological approaches for the management of acute and chronic conditions, like obesity and metabolic diseases, without disturbing the homeostatic functions of IL-6 and microglia in the hypothalamus.
Collapse
Affiliation(s)
- Vanessa C. D. Bobbo
- Faculty of Nursing, University of Campinas, SP 13083-887, Brazil
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, SP 13083-864, Brazil
| | - Carlos P. Jara
- Faculty of Nursing, University of Campinas, SP 13083-887, Brazil
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, SP 13083-864, Brazil
| | - Natália F. Mendes
- Faculty of Nursing, University of Campinas, SP 13083-887, Brazil
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, SP 13083-864, Brazil
| | - Joseane Morari
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, SP 13083-864, Brazil
| | - Lício A. Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, SP 13083-864, Brazil
| | - Eliana P. Araújo
- Faculty of Nursing, University of Campinas, SP 13083-887, Brazil
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, SP 13083-864, Brazil
| |
Collapse
|
29
|
Maternal n-3 PUFAs deficiency during pregnancy inhibits neural progenitor cell proliferation in fetal rat cerebral cortex. Int J Dev Neurosci 2019; 76:72-79. [PMID: 31299388 DOI: 10.1016/j.ijdevneu.2019.07.001] [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: 05/08/2019] [Revised: 06/26/2019] [Accepted: 07/05/2019] [Indexed: 11/20/2022] Open
Abstract
The aim of this study was to evaluate the in vivo impacts of maternal n-3 polyunsaturated fatty acids (PUFAs) deficiency during pregnancy on the proliferation of neural progenitor cells (NPCs) in the developing cerebral cortex of fetal rats. Our results showed that about 5 weeks of maternal dietary n-3 PUFAs deprivation resulted in a substantial n-3 PUFA deficiency in fetal rat cerebral cortex. Importantly, by two survival schemes and two quantitative methods, we found that maternal intake of n-3 PUFAs deficient diet during the gestation significantly inhibited the proliferation of NPCs in fetal rat cerebral cortex. Moreover, the decreased cortical NPCs proliferation induced by nutritional n-3 PUFAs restriction did not originate from the increased NPCs apoptosis. Finally, our observations indicated that the down-regulation of cyclin E protein might be involved in the inhibitory effects of maternal n-3 PUFAs deficient diet on the proliferation of cortical NPCs. These findings highlight the importance of maternal intake of appropriate n-3 PUFAs and deepen our understanding of the exact effects of n-3 PUFAs on mammalian brain development.
Collapse
|
30
|
Zhang TT, Xu J, Wang YM, Xue CH. Health benefits of dietary marine DHA/EPA-enriched glycerophospholipids. Prog Lipid Res 2019; 75:100997. [DOI: 10.1016/j.plipres.2019.100997] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 07/04/2019] [Accepted: 07/04/2019] [Indexed: 02/07/2023]
|
31
|
Leyrolle Q, Layé S, Nadjar A. Direct and indirect effects of lipids on microglia function. Neurosci Lett 2019; 708:134348. [PMID: 31238131 DOI: 10.1016/j.neulet.2019.134348] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 06/05/2019] [Accepted: 06/21/2019] [Indexed: 02/07/2023]
Abstract
Microglia are key players in brain function by maintaining brain homeostasis across lifetime. They participate to brain development and maturation through their ability to release neurotrophic factors, to remove immature synapses or unnecessary neural progenitors. They modulate neuronal activity in healthy adult brains and they also orchestrate the neuroinflammatory response in various pathophysiological contexts such as aging and neurodegenerative diseases. One of the main features of microglia is their high sensitivity to environmental factors, partly via the expression of a wide range of receptors. Recent data pinpoint that dietary fatty acids modulate microglia function. Both the quantity and the type of fatty acid are potent modulators of microglia physiology. The present review aims at dissecting the current knowledge on the direct and indirect mechanisms (focus on gut microbiota and hormones) through which fatty acids influence microglial physiology. We summarize main discoveries from in vitro and in vivo models on fatty acid-mediated microglial modulation. All these studies represent a promising field of research that could promote using nutrition as a novel therapeutic or preventive tool in diseases involving microglia dysfunctions.
Collapse
Affiliation(s)
- Q Leyrolle
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| | - S Layé
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| | - A Nadjar
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France.
| |
Collapse
|
32
|
Yam K, Schipper L, Reemst K, Ruigrok SR, Abbink MR, Hoeijmakers L, Naninck EFG, Zarekiani P, Oosting A, Van Der Beek EM, Lucassen PJ, Korosi A. Increasing availability of ω‐3 fatty acid in the early‐life diet prevents the early‐life stress‐induced cognitive impairments without affecting metabolic alterations. FASEB J 2019; 33:5729-5740. [DOI: 10.1096/fj.201802297r] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Kit‐Yi Yam
- Centre for NeuroscienceSwammerdam Institute for Life SciencesUniversity of Amsterdam Amsterdam The Netherlands
| | | | - Kitty Reemst
- Centre for NeuroscienceSwammerdam Institute for Life SciencesUniversity of Amsterdam Amsterdam The Netherlands
| | - Silvie R. Ruigrok
- Centre for NeuroscienceSwammerdam Institute for Life SciencesUniversity of Amsterdam Amsterdam The Netherlands
| | - Maralinde R. Abbink
- Centre for NeuroscienceSwammerdam Institute for Life SciencesUniversity of Amsterdam Amsterdam The Netherlands
| | - Lianne Hoeijmakers
- Centre for NeuroscienceSwammerdam Institute for Life SciencesUniversity of Amsterdam Amsterdam The Netherlands
| | - Eva F. G. Naninck
- Centre for NeuroscienceSwammerdam Institute for Life SciencesUniversity of Amsterdam Amsterdam The Netherlands
| | - Parand Zarekiani
- Centre for NeuroscienceSwammerdam Institute for Life SciencesUniversity of Amsterdam Amsterdam The Netherlands
| | | | - Eline M. Van Der Beek
- Danone Nutricia Research Utrecht The Netherlands
- Department of PediatricsUniversity Medical Centre GroningenUniversity of Groningen Groningen The Netherlands
| | - Paul J. Lucassen
- Centre for NeuroscienceSwammerdam Institute for Life SciencesUniversity of Amsterdam Amsterdam The Netherlands
| | - Aniko Korosi
- Centre for NeuroscienceSwammerdam Institute for Life SciencesUniversity of Amsterdam Amsterdam The Netherlands
| |
Collapse
|