1
|
Sun YX, Guo JB, Chen BL. Effects of Physical Exercise on High-Fat Diet-Induced Hypothalamic Inflammation: Systematic Review. Mol Neurobiol 2025:10.1007/s12035-025-04927-0. [PMID: 40220246 DOI: 10.1007/s12035-025-04927-0] [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: 11/26/2024] [Accepted: 04/05/2025] [Indexed: 04/14/2025]
Abstract
The high-fat diet (HFD)-induced hypothalamic inflammatory response is considered a common pathogenic basis for various metabolic diseases. Exercise can improve overall metabolic health, and it exerts a positive impact on local inflammation. The purpose of this systematic review was to evaluate existing evidence and analyze the effects of exercise intervention on the behavior, inflammatory factors, glial cells, and feeding neurons of animals with HFD-induced hypothalamic inflammation. We systematically searched PubMed, Scopus, Ebsco, EMBASE, and Web of Science databases from database establishment to July 2024 to identify all animal studies on exercise intervention in HFD-induced hypothalamic inflammation. Two researchers independently conducted literature screening to determine inclusion in the study. Finally, SYRCLE's risk of bias tool for animal studies was used to assess the risk of bias in the included studies. A total of 11 studies were included. Among them, 10 studies used mice and 1 study used rabbits. Overall, the inclusion of research supported the improvement of behavior, inflammatory factors, glial cells, and feeding neurons in animals with HFD-induced hypothalamic inflammation through exercise. However, 2 studies did not find significant changes in food intake and body weight, and 4 studies found that exercise intervention reduced the expression of anti-inflammatory factors (IL- 10 and SOCS3). Different exercise programs, such as forced and voluntary exercise, may be potential reasons for the differences in outcomes. On the basis of existing evidence, exercise appeared to be a potential protective and preventive factor against HFD-induced hypothalamic inflammation. However, the inconsistent results suggest that further research is needed in the future to thoroughly elucidate the mechanisms by which exercise improves and prevents hypothalamic inflammation.
Collapse
Affiliation(s)
- Yu Xue Sun
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Jia Bao Guo
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Bing Lin Chen
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
2
|
Wu G, Li F, Li Y, Li S, Alam MJ, Chen JDZ. Progressive impairment in gastric and duodenal slow waves and autonomic function during progression of type 2 diabetes in rats. Am J Physiol Gastrointest Liver Physiol 2025; 328:G386-G398. [PMID: 39993032 DOI: 10.1152/ajpgi.00278.2024] [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: 09/03/2024] [Revised: 10/06/2024] [Accepted: 02/11/2025] [Indexed: 02/26/2025]
Abstract
The abnormalities of gastrointestinal (GI) slow waves play key roles in the pathophysiology of diabetic gastroparesis, which is highly prevalent in type 2 diabetes (T2D). Although relatively well-investigated in diabetic enteric neuropathy, abnormalities and progressive impairments of gastric slow waves (GSWs) and duodenal slow waves (DSWs) are underinvestigated during the progression of T2D. The aim of this study was to explore alterations in GSW and DSW during the development of diabetes induced by high-fat diet (HFD) followed by a low dose of streptozotocin (STZ). Weekly recordings of slow waves from healthy, prediabetic to diabetes stages exhibited a progressively decreased percentage of normal slow waves (%NSW) starting after HFD feeding (prediabetic stage) in the fasting state and starting after STZ injection (diabetic stage) in the postprandial state. The postprandial increase in the power of slow waves observed in normal control rats was absent starting from 2 wk after HFD and persisted after STZ. The mechanism might be attributed to both progressively increased blood glucose (BG) and impaired autonomic function in view of the following results: 1) the %NSW was negatively correlated with the fasting BG; 2) during the oral glucose tolerance test, %NSW of DSW and BG exhibited a positive correlation in rats with hemoglobin A1C (HbA1C) < 5.0%, but a negative correlation in rats with HbA1C ≥ 5.0%; and 3) in comparison with baseline (healthy stage) of the same cohort, plasma pancreatic polypeptide (reflecting vagal activity) was progressively decreased, whereas plasma norepinephrine (reflecting sympathetic activity) was progressively increased.NEW & NOTEWORTHY This study recorded the progressive impairment in the regularity of gastric and duodenal slow waves in a rat model mimicking the progression to type 2 diabetes including the stage of health, prediabetic stage, and diabetes. The progressive impairment in gastric/duodenal slow waves might be attributed to the progressive increase in blood glucose and impairment in autonomic function.
Collapse
Affiliation(s)
- Gaojue Wu
- Division of Gastroenterology and Hepatology, School of Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Fei Li
- Division of Gastroenterology and Hepatology, School of Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Yan Li
- Division of Gastroenterology and Hepatology, School of Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Shiying Li
- Division of Gastroenterology and Hepatology, School of Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Md Jahangir Alam
- Division of Gastroenterology and Hepatology, School of Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Jiande D Z Chen
- Division of Gastroenterology and Hepatology, School of Medicine, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
3
|
Mendez-Hernandez R, Braga I, Bali A, Yang M, de Lartigue G. Vagal Sensory Gut-Brain Pathways That Control Eating-Satiety and Beyond. Compr Physiol 2025; 15:e70010. [PMID: 40229922 DOI: 10.1002/cph4.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025]
Abstract
The vagus nerve is the body's primary sensory conduit from gut to brain, traditionally viewed as a passive relay for satiety signals. However, emerging evidence reveals a far more complex system-one that actively encodes diverse aspects of meal-related information, from mechanical stretch to nutrient content, metabolic state, and even microbial metabolites. This review challenges the view of vagal afferent neurons (VANs) as simple meal-termination sensors and highlights their specialized subpopulations, diverse sensory modalities, and downstream brain circuits, which shape feeding behavior, metabolism, and cognition. We integrate recent advances from single-cell transcriptomics, neural circuit mapping, and functional imaging to examine how VANs contribute to gut-brain communication beyond satiety, including their roles in food reward and memory formation. By synthesizing the latest research and highlighting emerging directions for the field, this review provides a comprehensive update on vagal sensory pathways and their role as integrators of meal information.
Collapse
Affiliation(s)
- Rebeca Mendez-Hernandez
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Isadora Braga
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Avnika Bali
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mingxin Yang
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Guillaume de Lartigue
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
4
|
Gruber T, Lechner F, Krieger JP, García-Cáceres C. Neuroendocrine gut-brain signaling in obesity. Trends Endocrinol Metab 2025; 36:42-54. [PMID: 38821753 DOI: 10.1016/j.tem.2024.05.002] [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: 03/31/2024] [Revised: 04/29/2024] [Accepted: 05/03/2024] [Indexed: 06/02/2024]
Abstract
The past decades have witnessed the rise and fall of several, largely unsuccessful, therapeutic attempts to bring the escalating obesity pandemic to a halt. Looking back to look ahead, the field has now put its highest hopes in translating insights from how the gastrointestinal (GI) tract communicates with the brain to calibrate behavior, physiology, and metabolism. A major focus of this review is to summarize the latest advances in comprehending the neuroendocrine aspects of this so-called 'gut-brain axis' and to explore novel concepts, cutting-edge technologies, and recent paradigm-shifting experiments. These exciting insights continue to refine our understanding of gut-brain crosstalk and are poised to promote the development of additional therapeutic avenues at the dawn of a new era of antiobesity therapeutics.
Collapse
Affiliation(s)
- Tim Gruber
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49506, USA; Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49506, USA; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany.
| | - Franziska Lechner
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany
| | - Jean-Philippe Krieger
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland; Institute of Neuroscience and Physiology, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Cristina García-Cáceres
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, 80336 Munich, Germany.
| |
Collapse
|
5
|
Emmons HA, Fordahl SC. Moderate-intensity aerobic exercise enhanced dopamine signaling in diet-induced obese female mice without preventing body weight gain. Neuroscience 2024; 555:1-10. [PMID: 39032807 PMCID: PMC11344652 DOI: 10.1016/j.neuroscience.2024.07.020] [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: 02/28/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 07/23/2024]
Abstract
Obesity continues to rise in prevalence and financial burden despite strong evidence linking it to an increased risk of developing several chronic diseases. Dopamine response and receptor density are shown to decrease under conditions of obesity. However, it is unclear if this could be a potential mechanism for treatment without drugs that have a potential for abuse. Therefore, the aim of this study was to investigate whether moderate-intensity exercise could reduce body weight gain and the associated decreases in dopamine signaling observed with high-fat diet-induced adiposity. We hypothesized that exercise would attenuate body weight gain and diet-induced inflammation in high-fat (HF)-fed mice, resulting in dopamine signaling (release and reuptake rate) comparable to sedentary, low-fat (LF)-fed counterparts. This hypothesis was tested using a mouse model of diet-induced obesity (DIO) and fast-scan cyclic voltammetry to measure evoked dopamine release and reuptake rates. Although the exercise protocol employed in this study was not sufficient to prevent significant body weight gain, there was an enhancement of dopamine signaling observed in female mice fed a HF diet that underwent treadmill running. Additionally, aerobic treadmill exercise enhanced the sensitivity to amphetamine (AMPH) in this same group of exercised, HF-fed females. The estrous cycle might influence the ability of exercise to enhance dopamine signaling in females, an effect not observed in male groups. Further research into females by estrous cycle phase, in addition to determining the optimal intensity and duration of aerobic exercise, are logical next steps.
Collapse
Affiliation(s)
| | - Steve C Fordahl
- UNC Greensboro, Department of Nutrition, Greensboro NC, USA.
| |
Collapse
|
6
|
Cutugno G, Kyriakidou E, Nadjar A. Rethinking the role of microglia in obesity. Neuropharmacology 2024; 253:109951. [PMID: 38615749 DOI: 10.1016/j.neuropharm.2024.109951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/16/2024]
Abstract
Microglia are the macrophages of the central nervous system (CNS), implying their role in maintaining brain homeostasis. To achieve this, these cells are sensitive to a plethora of endogenous and exogenous signals, such as neuronal activity, cellular debris, hormones, and pathological patterns, among many others. More recent research suggests that microglia are highly responsive to nutrients and dietary variations. In this context, numerous studies have demonstrated their significant role in the development of obesity under calorie surfeit. Because many reviews already exist on this topic, we have chosen to present the state of our reflections on various concepts put forth in the literature, bringing a new perspective whenever possible. Our literature review focuses on studies conducted in the arcuate nucleus of the hypothalamus, a key structure in the control of food intake. Specifically, we present the recent data available on the modifications of microglial energy metabolism following the consumption of an obesogenic diet and their consequences on hypothalamic neuron activity. We also highlight the studies unraveling the mechanisms underlying obesity-related sexual dimorphism. The review concludes with a list of questions that remain to be addressed in the field to achieve a comprehensive understanding of the role of microglia in the regulation of body energy metabolism. This article is part of the Special Issue on "Microglia".
Collapse
Affiliation(s)
- G Cutugno
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France
| | - E Kyriakidou
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France
| | - A Nadjar
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France; Institut Universitaire de France (IUF), France.
| |
Collapse
|
7
|
Minaya DM, Kim JS, Kirkland R, Allen J, Cullinan S, Maclang N, de Lartigue G, de La Serre CB. Transfer with microbiota from lean donors prevents excessive weight gain and restores gut-brain vagal signaling in obese rats maintained on a high fat diet. RESEARCH SQUARE 2024:rs.3.rs-4438240. [PMID: 38853960 PMCID: PMC11160927 DOI: 10.21203/rs.3.rs-4438240/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Background The collection of microorganisms, mainly bacteria, which live in the gastrointestinal (GI) tract are collectible known as the gut microbiota. GI bacteria play an active role in regulation of the host's immune system and metabolism, as well as certain pathophysiological processes. Diet is the main factor modulating GI microbiota composition and recent studies have shown that high fat (HF) diets induce detrimental changes, known as dysbiosis, in the GI bacterial makeup. HF diet induced microbiota dysbiosis has been associated with structural and functional changes in gut-brain vagally mediated signaling system, associated with overeating and obesity. Although HF-driven changes in microbiota composition are sufficient to alter vagal signaling, it is unknown if restoring normal microbiota in obesity can improve gut-brain signaling and metabolic outcomes. In this study, we evaluated the effect of lean gut microbiota transfer in obese, vagally compromised, rats on gut-brain communication, food intake, and body weight. Male Sprague-Dawley rats were maintained on regular chow, or 45% HF diet for nine weeks followed by three weeks of microbiota depletion using an antibiotic cocktail. The animals were then divided into four groups (n=10 each): LF - control group on regular chow, LF-LF - chow fed animals that received antibiotics and microbiota from chow fed animals, HF-LF - HF fed animals that received microbiota from chow fed animals, and HF-HF - HF fed animals that received microbiota from HF fed animals. Animals were gavaged with donor microbiota for three consecutive days on week one and once a week thereafter for three more weeks. HF-LF animals received inulin as a prebiotic to aid the establishment of the lean microbiome. Results We found that transferring a LF microbiota to HF fed animals (HF-LF) reduced caloric intake during the light phase when compared with HF-HF rats and prevented additional excessive weight gain. We did not observe significant changes in the density of vagal afferents terminating in the brainstem among the groups, however, HF-LF animals displayed an increase in postprandial activation of both primary sensory neurons innervating the GI tract and brainstem secondary neurons. Conclusions We concluded from these data that normalizing microbiota composition in obese rats improves gut-brain communication and restores normal feeding patterns which was associated with a reduction in weight gain.
Collapse
Affiliation(s)
- Dulce M. Minaya
- Department of Nutritional Science, University of Georgia, Athens, GA
| | | | - Rebecca Kirkland
- Department of Nutritional Science, University of Georgia, Athens, GA
| | - Jillian Allen
- Department of Nutritional Science, University of Georgia, Athens, GA
| | - Sitara Cullinan
- Department of Nutritional Science, University of Georgia, Athens, GA
| | - Neil Maclang
- Department of Nutritional Science, University of Georgia, Athens, GA
| | | | | |
Collapse
|
8
|
Boutanquoi PM, Khan AS, Cabeza L, Jantzen L, Gautier T, Yesylevskyy S, Ramseyer C, Masson D, Van Waes V, Hichami A, Khan NA. A novel fatty acid analogue triggers CD36-GPR120 interaction and exerts anti-inflammatory action in endotoxemia. Cell Mol Life Sci 2024; 81:176. [PMID: 38598021 PMCID: PMC11006773 DOI: 10.1007/s00018-024-05207-1] [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: 01/30/2024] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 04/11/2024]
Abstract
Inflammation is a mediator of a number of chronic pathologies. We synthesized the diethyl (9Z,12Z)-octadeca-9,12-dien-1-ylphosphonate, called NKS3, which decreased lipopolysaccharide (LPS)-induced mRNA upregulation of proinflammatory cytokines (IL-1β, IL-6 and TNF-α) not only in primary intraperitoneal and lung alveolar macrophages, but also in freshly isolated mice lung slices. The in-silico studies suggested that NKS3, being CD36 agonist, will bind to GPR120. Co-immunoprecipitation and proximity ligation assays demonstrated that NKS3 induced protein-protein interaction of CD36 with GPR120in RAW 264.7 macrophage cell line. Furthermore, NKS3, via GPR120, decreased LPS-induced activation of TAB1/TAK1/JNK pathway and the LPS-induced mRNA expression of inflammatory markers in RAW 264.7 cells. In the acute lung injury model, NKS3 decreased lung fibrosis and inflammatory cytokines (IL-1β, IL-6 and TNF-α) and nitric oxide (NO) production in broncho-alveolar lavage fluid. NKS3 exerted a protective effect on LPS-induced remodeling of kidney and liver, and reduced circulating IL-1β, IL-6 and TNF-α concentrations. In a septic shock model, NKS3 gavage decreased significantly the LPS-induced mortality in mice. In the last, NKS3 decreased neuroinflammation in diet-induced obese mice. Altogether, these results suggest that NKS3 is a novel anti-inflammatory agent that could be used, in the future, for the treatment of inflammation-associated pathologies.
Collapse
Affiliation(s)
- Pierre-Marie Boutanquoi
- Physiologie de la Nutrition & Toxicologie, UMR U1231 INSERM/Université de Bourgogne/Agro-Sup, Université Bourgogne Franche-Comté, 6 Boulevard Gabriel, 21000, Dijon, France
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France
| | - Amira Sayed Khan
- Physiologie de la Nutrition & Toxicologie, UMR U1231 INSERM/Université de Bourgogne/Agro-Sup, Université Bourgogne Franche-Comté, 6 Boulevard Gabriel, 21000, Dijon, France
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France
| | - Lidia Cabeza
- Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive-UR LINC, UFC, Besançon, France
| | - Lucas Jantzen
- Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive-UR LINC, UFC, Besançon, France
| | - Thomas Gautier
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France
- LIPNESS, UMR U1231 INSERM/UB/Agro-Sup, Université Bourgogne Franche-Comté, 21000, Dijon, France
| | - Semen Yesylevskyy
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, 166 10, Prague 6, Czech Republic
- Laboratoire Chrono Environnement UMR CNRS6249, Université de Bourgogne Franche-Comté (UBFC), 16 route de Gray, 25030, Besançon, Cedex, France
- Receptor.AI Inc., 20-22 Wenlock Road, London, N1 7GU, UK
- Department of Physical Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 12, 771 46, Olomouc, Czech Republic
- Department of Physics of Biological Systems, Institute of Physics of the National Academy of Sciences of Ukraine, Prospect Nauky 46, Kiev, 03028, Ukraine
| | - Christophe Ramseyer
- Laboratoire Chrono Environnement UMR CNRS6249, Université de Bourgogne Franche-Comté (UBFC), 16 route de Gray, 25030, Besançon, Cedex, France
| | - David Masson
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France
- LIPNESS, UMR U1231 INSERM/UB/Agro-Sup, Université Bourgogne Franche-Comté, 21000, Dijon, France
| | - Vincent Van Waes
- Laboratoire de Recherches Intégratives en Neurosciences et Psychologie Cognitive-UR LINC, UFC, Besançon, France
| | - Aziz Hichami
- Physiologie de la Nutrition & Toxicologie, UMR U1231 INSERM/Université de Bourgogne/Agro-Sup, Université Bourgogne Franche-Comté, 6 Boulevard Gabriel, 21000, Dijon, France
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France
| | - Naim Akhtar Khan
- Physiologie de la Nutrition & Toxicologie, UMR U1231 INSERM/Université de Bourgogne/Agro-Sup, Université Bourgogne Franche-Comté, 6 Boulevard Gabriel, 21000, Dijon, France.
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France.
| |
Collapse
|
9
|
Başer Ö, Yavuz Y, Özen DÖ, Özgün HB, Ağuş S, Civaş CC, Atasoy D, Yılmaz B. Effects of chronic high fat diet on mediobasal hypothalamic satiety neuron function in POMC-Cre mice. Mol Metab 2024; 82:101904. [PMID: 38395148 PMCID: PMC10910127 DOI: 10.1016/j.molmet.2024.101904] [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: 11/30/2023] [Revised: 02/12/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024] Open
Abstract
OBJECTIVE The prevalence of obesity has increased over the past three decades. Proopiomelanocortin (POMC) neurons in the hypothalamic arcuate nucleus (ARC) play a vital role in induction of satiety. Chronic consumption of high-fat diet is known to reduce hypothalamic neuronal sensitivity to hormones like leptin, thus contributing to the development and persistence of obesity. The functional and morphological effects of a high-calorie diet on POMC neurons and how these effects contribute to the development and maintenance of the obese phenotype are not fully understood. For this purpose, POMC-Cre transgenic mice model was exposed to high-fat diet (HFD) and at the end of a 3- and 6-month period, electrophysiological and morphological changes, and the role of POMC neurons in homeostatic nutrition and their response to leptin were thoroughly investigated. METHODS Effects of HFD on POMC-satiety neurons in transgenic mice models exposed to chronic high-fat diet were investigated using electrophysiological (patch-clamp), chemogenetic and Cre recombinase advanced technological methods. Leptin, glucose and lipid profiles were determined and analyzed. RESULTS In mice exposed to a high-fat diet for 6 months, no significant changes in POMC dendritic spine number or projection density from POMC neurons to the paraventricular hypothalamus (PVN), lateral hypothalamus (LH), and bed nucleus stria terminalis (BNST) were observed. It was revealed that leptin hormone did not change the electrophysiological activities of POMC neurons in mice fed with HFD for 6 months. In addition, chemogenetic stimulation of POMC neurons increased HFD consumption. In the 3-month HFD-fed group, POMC activation induced an orexigenic response in mice, whereas switching to a standard diet was found to abolish orexigenic behavior in POMC mice. CONCLUSIONS Chronic high fat consumption disrupts the regulation of POMC neuron activation by leptin. Altered POMC neuron activation abolished the neuron's characteristic behavioral anorexigenic response. Change in nutritional content contributes to the reorganization of developing maladaptations.
Collapse
Affiliation(s)
- Özge Başer
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Yavuz Yavuz
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Deniz Öykü Özen
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Hüseyin Buğra Özgün
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Sami Ağuş
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Cihan Civan Civaş
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye
| | - Deniz Atasoy
- University of Iowa, Carver College of Medicine, Department of Neuroscience and Pharmacology, Iowa City, USA
| | - Bayram Yılmaz
- Yeditepe University, Faculty of Medicine, Department of Physiology, Istanbul, Türkiye; Izmir Biomedicine and Genome Center, Izmir, Türkiye.
| |
Collapse
|
10
|
Jantzen L, Dumontoy S, Ramadan B, Houdayer C, Haffen E, Hichami A, Khan NA, Van Waes V, Cabeza L. Dietary linoleic acid supplementation protects against obesity-induced microglial reactivity in mice. Sci Rep 2024; 14:6644. [PMID: 38503857 PMCID: PMC10951280 DOI: 10.1038/s41598-024-56959-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/13/2024] [Indexed: 03/21/2024] Open
Abstract
We investigated whether linoleic acid (LA) supplementation could modulate emotional behavior and microglia-related neuroinflammation. For that, male mice of C57BL/6J genetic background fed either a high-fat diet (HFD) or a standard diet (STD) for 12 weeks, were treated with a vehicle or LA solution for 5 weeks before being evaluated for emotional behavior using a battery of behavioral tests. The animals were subsequently sacrificed and their brains collected and processed for immunofluorescence staining, targeting microglia-specific calcium-binding proteins (IBA-1). Neuroinflammation severity was assessed in multiple hypothalamic, cortical and subcortical brain regions. We show an anxio-depressive-like effect of sustained HFD feeding that was neither alleviated nor worsened with LA supplementation. However, increased IBA-1 expression and microgliosis in the HFD group were largely attenuated by LA supplementation. These observations demonstrate that the anti-neuroinflammatory properties of LA are not restricted to hypothalamic areas but are also evident at the cortical and subcortical levels. This study discloses that neuroinflammation plays a role in the genesis of neuropsychiatric disorders in the context of obesity, and that LA supplementation is a useful dietary strategy to alleviate the impact of obesity-related neuroinflammation.
Collapse
Affiliation(s)
- Lucas Jantzen
- Université de Franche-Comté, UMR INSERM 1322 LINC, 19, Rue Ambroise Paré, 25000, Besançon Cedex, France
| | - Stéphanie Dumontoy
- Université de Franche-Comté, UMR INSERM 1322 LINC, 19, Rue Ambroise Paré, 25000, Besançon Cedex, France
| | - Bahrie Ramadan
- Université de Franche-Comté, UMR INSERM 1322 LINC, 19, Rue Ambroise Paré, 25000, Besançon Cedex, France
| | - Christophe Houdayer
- Université de Franche-Comté, UMR INSERM 1322 LINC, 19, Rue Ambroise Paré, 25000, Besançon Cedex, France
| | - Emmanuel Haffen
- Université de Franche-Comté, UMR INSERM 1322 LINC, service de psychiatrie de l'adulte, CIC-1431 INSERM, CHU de Besançon, 25030, Besançon, France
| | - Aziz Hichami
- Physiologie de la Nutrition & Toxicologie (NUTox), UMR UB/Institut Agro/INSERM U1231, Lipides, Nutrition & Cancer, LABEX-LipStick, Université de Bourgogne, Dijon, France
| | - Naim Akhtar Khan
- Physiologie de la Nutrition & Toxicologie (NUTox), UMR UB/Institut Agro/INSERM U1231, Lipides, Nutrition & Cancer, LABEX-LipStick, Université de Bourgogne, Dijon, France
| | - Vincent Van Waes
- Université de Franche-Comté, UMR INSERM 1322 LINC, 19, Rue Ambroise Paré, 25000, Besançon Cedex, France
| | - Lidia Cabeza
- Université de Franche-Comté, UMR INSERM 1322 LINC, 19, Rue Ambroise Paré, 25000, Besançon Cedex, France.
| |
Collapse
|
11
|
Salvi J, Andreoletti P, Audinat E, Balland E, Ben Fradj S, Cherkaoui-Malki M, Heurtaux T, Liénard F, Nédélec E, Rovère C, Savary S, Véjux A, Trompier D, Benani A. Microgliosis: a double-edged sword in the control of food intake. FEBS J 2024; 291:615-631. [PMID: 35880408 DOI: 10.1111/febs.16583] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/30/2022] [Accepted: 07/25/2022] [Indexed: 02/16/2024]
Abstract
Maintaining energy balance is essential for survival and health. This physiological function is controlled by the brain, which adapts food intake to energy needs. Indeed, the brain constantly receives a multitude of biological signals that are derived from digested foods or that originate from the gastrointestinal tract, energy stores (liver and adipose tissues) and other metabolically active organs (muscles). These signals, which include circulating nutrients, hormones and neuronal inputs from the periphery, collectively provide information on the overall energy status of the body. In the brain, several neuronal populations can specifically detect these signals. Nutrient-sensing neurons are found in discrete brain areas and are highly enriched in the hypothalamus. In turn, specialized brain circuits coordinate homeostatic responses acting mainly on appetite, peripheral metabolism, activity and arousal. Accumulating evidence shows that hypothalamic microglial cells located at the vicinity of these circuits can influence the brain control of energy balance. However, microglial cells could have opposite effects on energy balance, that is homeostatic or detrimental, and the conditions for this shift are not totally understood yet. One hypothesis relies on the extent of microglial activation, and nutritional lipids can considerably change it.
Collapse
Affiliation(s)
- Juliette Salvi
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Pierre Andreoletti
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Etienne Audinat
- IGF, Université de Montpellier, CNRS, Inserm, Montpellier, France
| | - Eglantine Balland
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Notting Hill, Australia
| | - Selma Ben Fradj
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Valbonne, France
| | | | - Tony Heurtaux
- Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg
- Department of Life Sciences and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Fabienne Liénard
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Emmanuelle Nédélec
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Carole Rovère
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Valbonne, France
| | - Stéphane Savary
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Anne Véjux
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Doriane Trompier
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Alexandre Benani
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
12
|
Minaya DM, Kim JS, Kirkland R, Allen J, Cullinan S, Maclang N, de Lartigue G, de La Serre C. Transfer of microbiota from lean donors in combination with prebiotics prevents excessive weight gain and improves gut-brain vagal signaling in obese rats. Gut Microbes 2024; 16:2421581. [PMID: 39485288 PMCID: PMC11540078 DOI: 10.1080/19490976.2024.2421581] [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: 06/20/2024] [Revised: 10/03/2024] [Accepted: 10/21/2024] [Indexed: 11/03/2024] Open
Abstract
Gastrointestinal (GI) microbiota plays an active role in regulating the host's immune system and metabolism, as well as certain pathophysiological processes. Diet is the main factor modulating GI microbiota composition and studies have shown that high fat (HF) diets induce detrimental changes (dysbiosis) in the GI bacterial makeup. HF diet induced dysbiosis has been associated with structural and functional changes in gut-brain vagally mediated signaling system, associated with overeating and obesity. Although HF-driven changes in microbiota composition are sufficient to alter vagal signaling, it is unknown if improving microbiota composition after diet-induced obesity has been established can ameliorate gut-brain signaling and metabolic outcomes. In this study, we evaluated the effect of lean gut microbiota transfer in obese, vagally compromised, rats on gut-brain communication, food intake, and body weight. Male rats were maintained on regular chow or 45% HF diet for nine weeks followed by three weeks of microbiota depletion using antibiotics. The animals were then divided into four groups (n = 10 each): LF - control fed regular chow, LF-LF - chow fed animals that received microbiota from chow fed donors, HF-LF - HF fed animals that received microbiota from chow fed donors, and HF-HF - HF fed animals that received microbiota from HF fed donors. HF-LF animals received inulin as a prebiotic to aid the establishment of the lean microbiome. We found that transferring a LF microbiota to HF fed animals (HF-LF) reduced caloric intake during the light phase when compared with HF-HF rats and prevented additional excessive weight gain. HF-LF animals displayed an increase in postprandial activation of both primary sensory neurons innervating the GI tract and brainstem secondary neurons. We concluded from these data that improving microbiota composition in obese rats is sufficient to ameliorate gut-brain communication and restore normal feeding patterns which was associated with a reduction in weight gain.
Collapse
Affiliation(s)
- Dulce M. Minaya
- Department of Nutritional Science, University of Georgia, Athens, GA, USA
| | - Jiyoung S Kim
- Emory University School of Medicine, Atlanta, GA, USA
| | - Rebecca Kirkland
- Department of Nutritional Science, University of Georgia, Athens, GA, USA
| | - Jillian Allen
- Department of Nutritional Science, University of Georgia, Athens, GA, USA
| | - Sitara Cullinan
- Department of Nutritional Science, University of Georgia, Athens, GA, USA
| | - Neil Maclang
- Department of Nutritional Science, University of Georgia, Athens, GA, USA
| | | | - Claire de La Serre
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
13
|
Lopes PKF, Costa SDO, Simino LADP, Chaves WF, Silva FA, Costa CL, Milanski M, Ignacio-Souza LM, Torsoni AS, Torsoni MA. Hypothalamic inflammation and the development of an obese phenotype induced by high-fat diet consumption is exacerbated in alpha7 nicotinic cholinergic receptor knockout mice. Food Res Int 2024; 176:113808. [PMID: 38163714 DOI: 10.1016/j.foodres.2023.113808] [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: 09/14/2023] [Revised: 11/27/2023] [Accepted: 12/02/2023] [Indexed: 01/03/2024]
Abstract
Hypothalamic inflammation and metabolic changes resulting from the consumption of high-fat diets have been linked to low grade inflammation and obesity. Inflammation impairs the hypothalamic expression of α7 nicotinic acetylcholine receptor (α7nAChR). The α7nAChR is described as the main component of the anti-inflammatory cholinergic pathway in different inflammation models. To assess whether the reduction in α7nAChR expression exacerbates hypothalamic inflammation induced by a high-fat diet (HFD), were used male and female global α7nAChR knockout mouse line in normal or high-fat diet for 4 weeks. Body weight gain, adiposity, glucose homeostasis, hypothalamic inflammation, food intake, and energy expenditure were evaluated. Insulin sensitivity was evaluated in neuronal cell culture. Consumption of an HFD for 4 weeks resulted in body weight gain and adiposity in male Chrna7-/- mice and the hypothalamus of male Chrna7-/- mice showed neuroinflammatory markers, with increased gene expression of pro-inflammatory cytokines and dysregulation in the nuclear factor kappa B pathway. Moreover, male Chrna7-/- mice consuming an HFD showed alterations in glucose homeostasis and serum of Chrna7-/- mice that consumed an HFD impaired insulin signalling in neuronal cell culture experiments. In general, female Chrna7-/- mice that consumed an HFD did not show the phenotypic and molecular changes found in male mice, indicating that there is sexual dimorphism in the analysed parameters. Thus, receptor deletion resulted in increased susceptibility to hypothalamic inflammation and metabolic damage associated with HFD consumption in male mice.
Collapse
Affiliation(s)
| | - Suleyma de Oliveira Costa
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil
| | - Laís A de Paula Simino
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil
| | - Wenicios Ferreira Chaves
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil
| | - Franciely Alves Silva
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil
| | - Caroline Lobo Costa
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil
| | - Marciane Milanski
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil; Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Brazil
| | - Leticia Martins Ignacio-Souza
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil; Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Brazil
| | - Adriana Souza Torsoni
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil; Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Brazil
| | - Marcio Alberto Torsoni
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil; Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Brazil.
| |
Collapse
|
14
|
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
|
15
|
Cozma D, Siatra P, Bornstein SR, Steenblock C. Sensitivity of the Neuroendocrine Stress Axis in Metabolic Diseases. Horm Metab Res 2024; 56:65-77. [PMID: 38171373 DOI: 10.1055/a-2201-6641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Metabolic diseases are prevalent in modern society and have reached pandemic proportions. Metabolic diseases have systemic effects on the body and can lead to changes in the neuroendocrine stress axis, the critical regulator of the body's stress response. These changes may be attributed to rising insulin levels and the release of adipokines and inflammatory cytokines by adipose tissue, which affect hormone production by the neuroendocrine stress axis. Chronic stress due to inflammation may exacerbate these effects. The increased sensitivity of the neuroendocrine stress axis may be responsible for the development of metabolic syndrome, providing a possible explanation for the high prevalence of severe comorbidities such as heart disease and stroke associated with metabolic disease. In this review, we address current knowledge of the neuroendocrine stress axis in response to metabolic disease and discuss its role in developing metabolic syndrome.
Collapse
Affiliation(s)
- Diana Cozma
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Panagiota Siatra
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Stefan R Bornstein
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
| | - Charlotte Steenblock
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
16
|
Custers E, Franco A, Kiliaan AJ. Bariatric Surgery and Gut-Brain-Axis Driven Alterations in Cognition and Inflammation. J Inflamm Res 2023; 16:5495-5514. [PMID: 38026245 PMCID: PMC10676679 DOI: 10.2147/jir.s437156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Obesity is associated with systemic inflammation, comorbidities like diabetes, cardiovascular disease and several cancers, cognitive decline and structural and functional brain changes. To treat, or potentially prevent these related comorbidities, individuals with obesity must achieve long-term sustainable weight loss. Often life style interventions, such as dieting and increased physical activity are not successful in achieving long-term weight loss. Meanwhile bariatric surgery has emerged as a safe and effective procedure to treat obesity. Bariatric surgery causes changes in physiological processes, but it is still not fully understood which exact mechanisms are involved. The successful weight loss after bariatric surgery might depend on changes in various energy regulating hormones, such as ghrelin, glucagon-like peptide-1 and peptide YY. Moreover, changes in microbiota composition and white adipose tissue functionality might play a role. Here, we review the effect of obesity on neuroendocrine effects, microbiota composition and adipose tissue and how these may affect inflammation, brain structure and cognition. Finally, we will discuss how these obesity-related changes may improve after bariatric surgery.
Collapse
Affiliation(s)
- Emma Custers
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, the Netherlands
| | - Ayla Franco
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, the Netherlands
| | - Amanda Johanne Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, the Netherlands
| |
Collapse
|
17
|
He Y, Yang K, Zhang L, Zhan M, Xia XW, Wang HF, Xie Y, Huang L, Yang N, Zheng YL, Yang H, Ying-Ning, Sun JY, Yang YJ, Ding WJ. Electroacupuncture for weight loss by regulating microglial polarization in the arcuate nucleus of the hypothalamus. Life Sci 2023; 330:121981. [PMID: 37516430 DOI: 10.1016/j.lfs.2023.121981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 07/31/2023]
Abstract
Electroacupuncture (EA) has a weight loss effect, but the underlying molecular mechanisms of weight loss with EA have not been fully elucidated. This study aimed to investigate the modulatory effects of EA on the phenotype of hypothalamic microglia in obese mice. A total of 50 male C57BL/6J mice were used in this study. There were three groups in this experiment: The conventional diet group (Chow group), the high-fat diet group (HFD group), and the EA intervention group (HFD + EA group). EA was applied at "Tianshu (ST25)", "Guanyuan (RN4)", "Zusanli (ST36)" and "Zhongwan (RN12)" every day for 10 min. Hematoxylin and eosin (H&E) staining, immunohistochemical staining, and real-time PCR were applied in this study. The results showed that EA intervention was associated with a decrease in body weight, food intake, adipose tissue weight, and adipocyte size. At the same time, EA induced microglia to exhibit an M2 phenotype, representing reduced iNOS/TNF-α and increased Arg-1/IL-10/BDNF, which may be due to the promotion of TREM2 expression. EA also reduced microglia enrichment in the hypothalamic arcuate nucleus and declined TLR4 and IL-6, inhibiting microglia-mediated neuroinflammation. In addition, EA treatment promoted POMC expression, which may be associated with reduced food intake and weight loss in obese mice. This work provides novel evidence of EA against obesity. However, further study is necessary of EA as a therapy for obesity.
Collapse
Affiliation(s)
- Yan He
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu, Sichuan 611137, China
| | - Kun Yang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu, Sichuan 611137, China
| | - Lu Zhang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu, Sichuan 611137, China
| | - Meng Zhan
- Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu, Sichuan 611137, China
| | - Xiu-Wen Xia
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu, Sichuan 611137, China
| | - Huai-Fu Wang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu, Sichuan 611137, China
| | - Ya Xie
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu, Sichuan 611137, China
| | - Ling Huang
- Hospital of Traditional Chinese Medicine, Yibin, Sichuan 644000, China
| | - Ni Yang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu, Sichuan 611137, China
| | - Ya-Li Zheng
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu, Sichuan 611137, China
| | - Hong Yang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu, Sichuan 611137, China
| | - Ying-Ning
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu, Sichuan 611137, China
| | - Jia-Yi Sun
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu, Sichuan 611137, China
| | - You-Jun Yang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu, Sichuan 611137, China.
| | - Wei-Jun Ding
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu, Sichuan 611137, China.
| |
Collapse
|
18
|
Azuma N, Mawatari T, Saito Y, Tsukamoto M, Sampei M, Iwama Y. Effect of Continuous Ingestion of Bifidobacteria and Dietary Fiber on Improvement in Cognitive Function: A Randomized, Double-Blind, Placebo-Controlled Trial. Nutrients 2023; 15:4175. [PMID: 37836458 PMCID: PMC10574581 DOI: 10.3390/nu15194175] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Bifidobacterium animalis subsp. lactis GCL2505 has been shown to have some positive effects on health, including improved defecation frequency and reduced visceral fat. These effects are thought to be due to GCL2505's unique ability to reach the intestine in a viable form and proliferate after a single intake. This leads to an increased number of intestinal bifidobacteria. This randomized, double-blind, placebo-controlled, parallel-group study was conducted to confirm that intake of GCL2505 and inulin (a prebiotic) improve cognitive function (n = 80). Participants consumed test drinks containing 1 × 1010 colony-forming units of GCL2505 per 100 g and 2.0 g of inulin per 100 g for 12 weeks. The change in cognitive function assessment scores was set as the primary endpoint. There were significant improvements in scores in the neurocognitive index domain, which is an assessment of overall cognitive function, in addition to overall attention, cognitive flexibility, and executive function domains. The intervention significantly increased the number of fecal bifidobacteria and affected the levels of several inflammatory markers. These results suggest that intake of GCL2505 and inulin improves cognitive function by improving the intestinal environment and alleviating inflammation.
Collapse
Affiliation(s)
- Naoki Azuma
- R&D Laboratory, Ezaki Glico Co., Ltd., 4-6-5, Utajima, Nishiyodogawa-Ku, Osaka 555-8502, Japan; (T.M.); (Y.S.); (M.T.); (M.S.)
| | - Takashi Mawatari
- R&D Laboratory, Ezaki Glico Co., Ltd., 4-6-5, Utajima, Nishiyodogawa-Ku, Osaka 555-8502, Japan; (T.M.); (Y.S.); (M.T.); (M.S.)
| | - Yasuo Saito
- R&D Laboratory, Ezaki Glico Co., Ltd., 4-6-5, Utajima, Nishiyodogawa-Ku, Osaka 555-8502, Japan; (T.M.); (Y.S.); (M.T.); (M.S.)
| | - Masashi Tsukamoto
- R&D Laboratory, Ezaki Glico Co., Ltd., 4-6-5, Utajima, Nishiyodogawa-Ku, Osaka 555-8502, Japan; (T.M.); (Y.S.); (M.T.); (M.S.)
| | - Masatoshi Sampei
- R&D Laboratory, Ezaki Glico Co., Ltd., 4-6-5, Utajima, Nishiyodogawa-Ku, Osaka 555-8502, Japan; (T.M.); (Y.S.); (M.T.); (M.S.)
| | - Yoshitaka Iwama
- Nihonbashi Cardiology Clinic, Kyodo Bldg. #201, 13-4 Nihonbashi Kodenmacho, Chuo-Ku, Tokyo 103-0001, Japan;
| |
Collapse
|
19
|
Fu X, Wang Y, Zhao F, Cui R, Xie W, Liu Q, Yang W. Shared biological mechanisms of depression and obesity: focus on adipokines and lipokines. Aging (Albany NY) 2023; 15:5917-5950. [PMID: 37387537 PMCID: PMC10333059 DOI: 10.18632/aging.204847] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/01/2023] [Indexed: 07/01/2023]
Abstract
Depression and obesity are both common disorders currently affecting public health, frequently occurring simultaneously within individuals, and the relationship between these disorders is bidirectional. The association between obesity and depression is highly co-morbid and tends to significantly exacerbate metabolic and related depressive symptoms. However, the neural mechanism under the mutual control of obesity and depression is largely inscrutable. This review focuses particularly on alterations in systems that may mechanistically explain the in vivo homeostatic regulation of the obesity and depression link, such as immune-inflammatory activation, gut microbiota, neuroplasticity, HPA axis dysregulation as well as neuroendocrine regulators of energy metabolism including adipocytokines and lipokines. In addition, the review summarizes potential and future treatments for obesity and depression and raises several questions that need to be answered in future research. This review will provide a comprehensive description and localization of the biological connection between obesity and depression to better understand the co-morbidity of obesity and depression.
Collapse
Affiliation(s)
- Xiying Fu
- Department of Endocrinology, The Second Hospital of Jilin University, Changchun 130041, P.R. China
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Yicun Wang
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Fangyi Zhao
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Ranji Cui
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Wei Xie
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Qianqian Liu
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Wei Yang
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
- Department of Neurology, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| |
Collapse
|
20
|
Escobar AP, Bonansco C, Cruz G, Dagnino-Subiabre A, Fuenzalida M, Negrón I, Sotomayor-Zárate R, Martínez-Pinto J, Jorquera G. Central and Peripheral Inflammation: A Common Factor Causing Addictive and Neurological Disorders and Aging-Related Pathologies. Int J Mol Sci 2023; 24:10083. [PMID: 37373230 PMCID: PMC10298583 DOI: 10.3390/ijms241210083] [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: 05/16/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Many diseases and degenerative processes affecting the nervous system and peripheral organs trigger the activation of inflammatory cascades. Inflammation can be triggered by different environmental conditions or risk factors, including drug and food addiction, stress, and aging, among others. Several pieces of evidence show that the modern lifestyle and, more recently, the confinement associated with the COVID-19 pandemic have contributed to increasing the incidence of addictive and neuropsychiatric disorders, plus cardiometabolic diseases. Here, we gather evidence on how some of these risk factors are implicated in activating central and peripheral inflammation contributing to some neuropathologies and behaviors associated with poor health. We discuss the current understanding of the cellular and molecular mechanisms involved in the generation of inflammation and how these processes occur in different cells and tissues to promote ill health and diseases. Concomitantly, we discuss how some pathology-associated and addictive behaviors contribute to worsening these inflammation mechanisms, leading to a vicious cycle that promotes disease progression. Finally, we list some drugs targeting inflammation-related pathways that may have beneficial effects on the pathological processes associated with addictive, mental, and cardiometabolic illnesses.
Collapse
Affiliation(s)
- Angélica P. Escobar
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.P.E.); (C.B.); (G.C.); (M.F.); (I.N.); (R.S.-Z.)
| | - Christian Bonansco
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.P.E.); (C.B.); (G.C.); (M.F.); (I.N.); (R.S.-Z.)
| | - Gonzalo Cruz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.P.E.); (C.B.); (G.C.); (M.F.); (I.N.); (R.S.-Z.)
| | - Alexies Dagnino-Subiabre
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.P.E.); (C.B.); (G.C.); (M.F.); (I.N.); (R.S.-Z.)
| | - Marco Fuenzalida
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.P.E.); (C.B.); (G.C.); (M.F.); (I.N.); (R.S.-Z.)
| | - Ignacio Negrón
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.P.E.); (C.B.); (G.C.); (M.F.); (I.N.); (R.S.-Z.)
| | - Ramón Sotomayor-Zárate
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.P.E.); (C.B.); (G.C.); (M.F.); (I.N.); (R.S.-Z.)
| | - Jonathan Martínez-Pinto
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.P.E.); (C.B.); (G.C.); (M.F.); (I.N.); (R.S.-Z.)
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Gonzalo Jorquera
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.P.E.); (C.B.); (G.C.); (M.F.); (I.N.); (R.S.-Z.)
- Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago 7830490, Chile
| |
Collapse
|
21
|
Müller L, Power Guerra N, Schildt A, Lindner T, Stenzel J, Behrangi N, Bergner C, Alberts T, Bühler D, Kurth J, Krause BJ, Janowitz D, Teipel S, Vollmar B, Kuhla A. [ 18F]GE-180-PET and Post Mortem Marker Characteristics of Long-Term High-Fat-Diet-Induced Chronic Neuroinflammation in Mice. Biomolecules 2023; 13:biom13050769. [PMID: 37238638 DOI: 10.3390/biom13050769] [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: 03/15/2023] [Revised: 04/14/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Obesity is characterized by immoderate fat accumulation leading to an elevated risk of neurodegenerative disorders, along with a host of metabolic disturbances. Chronic neuroinflammation is a main factor linking obesity and the propensity for neurodegenerative disorders. To determine the cerebrometabolic effects of diet-induced obesity (DIO) in female mice fed a long-term (24 weeks) high-fat diet (HFD, 60% fat) compared to a group on a control diet (CD, 20% fat), we used in vivo PET imaging with the radiotracer [18F]FDG as a marker for brain glucose metabolism. In addition, we determined the effects of DIO on cerebral neuroinflammation using translocator protein 18 kDa (TSPO)-sensitive PET imaging with [18F]GE-180. Finally, we performed complementary post mortem histological and biochemical analyses of TSPO and further microglial (Iba1, TMEM119) and astroglial (GFAP) markers as well as cerebral expression analyses of cytokines (e.g., Interleukin (IL)-1β). We showed the development of a peripheral DIO phenotype, characterized by increased body weight, visceral fat, free triglycerides and leptin in plasma, as well as increased fasted blood glucose levels. Furthermore, we found obesity-associated hypermetabolic changes in brain glucose metabolism in the HFD group. Our main findings with respect to neuroinflammation were that neither [18F]GE-180 PET nor histological analyses of brain samples seem fit to detect the predicted cerebral inflammation response, despite clear evidence of perturbed brain metabolism along with elevated IL-1β expression. These results could be interpreted as a metabolically activated state in brain-resident immune cells due to a long-term HFD.
Collapse
Affiliation(s)
- Luisa Müller
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, 18057 Rostock, Germany
- Department of Psychosomatic Medicine and Psychotherapy, Rostock University Medical Centre, 18147 Rostock, Germany
- Centre for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Centre, 18147 Rostock, Germany
| | - Nicole Power Guerra
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, 18057 Rostock, Germany
- Institute of Anatomy, Rostock University Medical Centre, 18057 Rostock, Germany
- Smell & Taste Clinic, Department of Otorhinolaryngology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01034 Dresden, Germany
| | - Anna Schildt
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Tobias Lindner
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Jan Stenzel
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Newshan Behrangi
- Institute of Anatomy and Cell Biology, Medical University of Bonn, 53115 Bonn, Germany
| | - Carina Bergner
- Department of Clinic and Polyclinic for Nuclear Medicine, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Teresa Alberts
- Institute of Anatomy and Cell Biology, Medical University of Bonn, 53115 Bonn, Germany
| | - Daniel Bühler
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Jens Kurth
- Department of Clinic and Polyclinic for Nuclear Medicine, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Bernd Joachim Krause
- Department of Clinic and Polyclinic for Nuclear Medicine, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Deborah Janowitz
- Department of Psychiatry, University of Greifswald, 17475 Greifswald, Germany
| | - Stefan Teipel
- Department of Psychosomatic Medicine and Psychotherapy, Rostock University Medical Centre, 18147 Rostock, Germany
- Centre for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Centre, 18147 Rostock, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock/Greifswald, 18147 Rostock, Germany
| | - Brigitte Vollmar
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, 18057 Rostock, Germany
- Centre for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Centre, 18147 Rostock, Germany
| | - Angela Kuhla
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, 18057 Rostock, Germany
- Centre for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Centre, 18147 Rostock, Germany
| |
Collapse
|
22
|
Vargas-Soria M, García-Alloza M, Corraliza-Gómez M. Effects of diabetes on microglial physiology: a systematic review of in vitro, preclinical and clinical studies. J Neuroinflammation 2023; 20:57. [PMID: 36869375 PMCID: PMC9983227 DOI: 10.1186/s12974-023-02740-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/16/2023] [Indexed: 03/05/2023] Open
Abstract
Diabetes mellitus is a heterogeneous chronic metabolic disorder characterized by the presence of hyperglycemia, commonly preceded by a prediabetic state. The excess of blood glucose can damage multiple organs, including the brain. In fact, cognitive decline and dementia are increasingly being recognized as important comorbidities of diabetes. Despite the largely consistent link between diabetes and dementia, the underlying causes of neurodegeneration in diabetic patients remain to be elucidated. A common factor for almost all neurological disorders is neuroinflammation, a complex inflammatory process in the central nervous system for the most part orchestrated by microglial cells, the main representatives of the immune system in the brain. In this context, our research question aimed to understand how diabetes affects brain and/or retinal microglia physiology. We conducted a systematic search in PubMed and Web of Science to identify research items addressing the effects of diabetes on microglial phenotypic modulation, including critical neuroinflammatory mediators and their pathways. The literature search yielded 1327 records, including 18 patents. Based on the title and abstracts, 830 papers were screened from which 250 primary research papers met the eligibility criteria (original research articles with patients or with a strict diabetes model without comorbidities, that included direct data about microglia in the brain or retina), and 17 additional research papers were included through forward and backward citations, resulting in a total of 267 primary research articles included in the scoping systematic review. We reviewed all primary publications investigating the effects of diabetes and/or its main pathophysiological traits on microglia, including in vitro studies, preclinical models of diabetes and clinical studies on diabetic patients. Although a strict classification of microglia remains elusive given their capacity to adapt to the environment and their morphological, ultrastructural and molecular dynamism, diabetes modulates microglial phenotypic states, triggering specific responses that include upregulation of activity markers (such as Iba1, CD11b, CD68, MHC-II and F4/80), morphological shift to amoeboid shape, secretion of a wide variety of cytokines and chemokines, metabolic reprogramming and generalized increase of oxidative stress. Pathways commonly activated by diabetes-related conditions include NF-κB, NLRP3 inflammasome, fractalkine/CX3CR1, MAPKs, AGEs/RAGE and Akt/mTOR. Altogether, the detailed portrait of complex interactions between diabetes and microglia physiology presented here can be regarded as an important starting point for future research focused on the microglia-metabolism interface.
Collapse
Affiliation(s)
- María Vargas-Soria
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain.,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain
| | - Mónica García-Alloza
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain.,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain
| | - Miriam Corraliza-Gómez
- Division of Physiology, School of Medicine, Universidad de Cadiz, Cadiz, Spain. .,Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), Cadiz, Spain.
| |
Collapse
|
23
|
Binayi F, Moslemi M, Khodagholi F, Hedayati M, Zardooz H. Long-term high-fat diet disrupts lipid metabolism and causes inflammation in adult male rats: possible intervention of endoplasmic reticulum stress. Arch Physiol Biochem 2023; 129:204-212. [PMID: 32907408 DOI: 10.1080/13813455.2020.1808997] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This study investigated the effect of long-term high-fat diet (HFD) on plasma lipid profile and probability of inflammation in adult rats. After weaning, male offspring were divided into six groups based on diet type and medication. After 20 weeks of dietary intake, 4-PBA (endoplasmic reticulum (ER) stress inhibitor) was injected for three days. Then, blood samples were taken to measure plasma concentrations of low-density lipoprotein (LDL), triglyceride (TG), high-density lipoprotein (HDL), cholesterol, leptin and interleukin 1-β (IL 1-β). The HFD increased body weight and food intake and intra-abdominal fat and thymus weights, which were associated with elevated plasma leptin level. Moreover, HFD increased plasma concentrations of TG, LDL, cholesterol and IL 1-β and decreased HDL level. Injection of 4-PBA reversed the plasma parameters changes caused by HFD. It seems that long-term HFD feeding through inducing the ER stress, disrupted the lipid metabolism and resulted in inflammation.
Collapse
Affiliation(s)
- Fateme Binayi
- Department of Physiology, School of Medicine, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Moslemi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homeira Zardooz
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Clyburn C, Carson KE, Smith CR, Travagli RA, Browning KN. Brainstem astrocytes control homeostatic regulation of caloric intake. J Physiol 2023; 601:801-829. [PMID: 36696965 PMCID: PMC10026361 DOI: 10.1113/jp283566] [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] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 12/08/2022] [Indexed: 01/27/2023] Open
Abstract
Prolonged high-fat diet (HFD) exposure is associated with hyperphagia, excess caloric intake and weight gain. After initial exposure to a HFD, a brief (24-48 h) period of hyperphagia is followed by the regulation of caloric intake and restoration of energy balance within an acute (3-5 day) period. Previous studies have demonstrated this occurs via a vagally mediated signalling cascade that increases glutamatergic transmission via activation of NMDA receptors located on gastric-projecting neurons of the dorsal motor nucleus of the vagus (DMV). The present study used electrophysiological recordings from thin brainstem slice preparations, in vivo recordings of gastric motility and tone, measurement of gastric emptying rates, and food intake studies to investigate the hypothesis that activation of brainstem astrocytes in response to acute HFD exposure is responsible for the increased glutamatergic drive to DMV neurons and the restoration of caloric balance. Pharmacological and chemogenetic inhibition of brainstem astrocytes reduced glutamatergic signalling and DMV excitability, dysregulated gastric tone and motility, attenuated the homeostatic delay in gastric emptying, and prevented the decrease in food intake that is observed during the period of energy regulation following initial exposure to HFD. Understanding the mechanisms involved in caloric regulation may provide critical insights into energy balance as well as into the hyperphagia that develops as these mechanisms are overcome. KEY POINTS: Initial exposure to a high fat diet is associated with a brief period of hyperphagia before caloric intake and energy balance is restored. This period of homeostatic regulation is associated with a vagally mediated signalling cascade that increases glutamatergic transmission to dorsal motor nucleus of the vagus (DMV) neurons via activation of synaptic NMDA receptors. The present study demonstrates that pharmacological and chemogenetic inhibition of brainstem astrocytes reduced glutamatergic signalling and DMV neuronal excitability, dysregulated gastric motility and tone and emptying, and prevented the regulation of food intake following high-fat diet exposure. Astrocyte regulation of glutamatergic transmission to DMV neurons appears to involve release of the gliotransmitters glutamate and ATP. Understanding the mechanisms involved in caloric regulation may provide critical insights into energy balance as well as into the hyperphagia that develops as these mechanisms are overcome.
Collapse
Affiliation(s)
- Courtney Clyburn
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
- Current position: Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, 97056
| | - Kaitlin E. Carson
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
| | - Caleb R. Smith
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
| | - R. Alberto Travagli
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
- Current position: Neurobiology Research, Newport, NC 28570
| | - Kirsteen N. Browning
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
| |
Collapse
|
25
|
Leptin Increases: Physiological Roles in the Control of Sympathetic Nerve Activity, Energy Balance, and the Hypothalamic-Pituitary-Thyroid Axis. Int J Mol Sci 2023; 24:ijms24032684. [PMID: 36769012 PMCID: PMC9917048 DOI: 10.3390/ijms24032684] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/17/2023] [Accepted: 01/21/2023] [Indexed: 02/04/2023] Open
Abstract
It is well established that decreases in plasma leptin levels, as with fasting, signal starvation and elicit appropriate physiological responses, such as increasing the drive to eat and decreasing energy expenditure. These responses are mediated largely by suppression of the actions of leptin in the hypothalamus, most notably on arcuate nucleus (ArcN) orexigenic neuropeptide Y neurons and anorexic pro-opiomelanocortin neurons. However, the question addressed in this review is whether the effects of increased leptin levels are also significant on the long-term control of energy balance, despite conventional wisdom to the contrary. We focus on leptin's actions (in both lean and obese individuals) to decrease food intake, increase sympathetic nerve activity, and support the hypothalamic-pituitary-thyroid axis, with particular attention to sex differences. We also elaborate on obesity-induced inflammation and its role in the altered actions of leptin during obesity.
Collapse
|
26
|
Marcos JL, Olivares-Barraza R, Ceballo K, Wastavino M, Ortiz V, Riquelme J, Martínez-Pinto J, Muñoz P, Cruz G, Sotomayor-Zárate R. Obesogenic Diet-Induced Neuroinflammation: A Pathological Link between Hedonic and Homeostatic Control of Food Intake. Int J Mol Sci 2023; 24:ijms24021468. [PMID: 36674982 PMCID: PMC9866213 DOI: 10.3390/ijms24021468] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 01/13/2023] Open
Abstract
Obesity-induced neuroinflammation is a chronic aseptic central nervous system inflammation that presents systemic characteristics associated with increased pro-inflammatory cytokines such as interleukin 1 beta (IL-1β) and interleukin 18 (IL-18) and the presence of microglia and reactive astrogliosis as well as the activation of the NLRP3 inflammasome. The obesity pandemic is associated with lifestyle changes, including an excessive intake of obesogenic foods and decreased physical activity. Brain areas such as the lateral hypothalamus (LH), lateral septum (LS), ventral tegmental area (VTA), and nucleus accumbens (NAcc) have been implicated in the homeostatic and hedonic control of feeding in experimental models of diet-induced obesity. In this context, a chronic lipid intake triggers neuroinflammation in several brain regions such as the hypothalamus, hippocampus, and amygdala. This review aims to present the background defining the significant impact of neuroinflammation and how this, when induced by an obesogenic diet, can affect feeding control, triggering metabolic and neurological alterations.
Collapse
Affiliation(s)
- José Luis Marcos
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Escuela de Ciencias Agrícolas y Veterinarias, Universidad Viña del Mar, Viña del Mar 2572007, Chile
- Programa de Doctorado en Ciencias e Ingeniería para la Salud, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Rossy Olivares-Barraza
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Programa de Doctorado en Ciencias Mención Neurociencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Karina Ceballo
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Programa de Doctorado en Ciencias Mención Neurociencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Melisa Wastavino
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Víctor Ortiz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Julio Riquelme
- Escuela de Medicina y Centro de Neurología Traslacional (CENTRAS), Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2540064, Chile
| | - Jonathan Martínez-Pinto
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Pablo Muñoz
- Escuela de Medicina y Centro de Neurología Traslacional (CENTRAS), Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2540064, Chile
| | - Gonzalo Cruz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Ramón Sotomayor-Zárate
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Correspondence: ; Tel.: +56-32-2508050
| |
Collapse
|
27
|
Smith DC, Karahan H, Wijeratne HRS, Al-Amin M, McCord B, Moon Y, Kim J. Deletion of the Alzheimer's disease risk gene Abi3 locus results in obesity and systemic metabolic disruption in mice. Front Aging Neurosci 2022; 14:1035572. [PMID: 36620768 PMCID: PMC9813750 DOI: 10.3389/fnagi.2022.1035572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
Alzheimer's disease (AD) genetics studies have identified a coding variant within ABI3 gene that increases the risk of developing AD. Recently, we demonstrated that deletion of the Abi3 gene locus dramatically exacerbates AD neuropathology in a transgenic mouse model of amyloidosis. In the course of this AD project, we unexpectedly found that deletion of the Abi3 gene locus resulted in a dramatic obese phenotype in non-transgenic mice. Here, we report our investigation into this serendipitous metabolic finding. Specifically, we demonstrate that mice with deletion of the Abi3 gene locus (Abi3-/- ) have dramatically increased body weight and body fat. Further, we determined that Abi3-/- mice have impaired energy expenditure. Additionally, we found that deletion of the Abi3 gene locus altered gene expression within the hypothalamus, particularly within immune-related pathways. Subsequent immunohistological analysis of the central nervous system (CNS) revealed that microglia number and area were decreased specifically within the mediobasal hypothalamus of Abi3-/- mice. Altogether, this investigation establishes the functional importance of the Abi3 gene locus in the regulation of systemic metabolism and maintenance of healthy body weight. While our previous findings indicated the importance of Abi3 in neurodegeneration, this study indicates that Abi3 related functions are also essential for metabolic regulation.
Collapse
Affiliation(s)
- Daniel C. Smith
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, United States
- Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Hande Karahan
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - H. R. Sagara Wijeratne
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Mamun Al-Amin
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Brianne McCord
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Younghye Moon
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jungsu Kim
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
28
|
Farkas GJ, Sneij A, McMillan DW, Tiozzo E, Nash MS, Gater DR. Energy expenditure and nutrient intake after spinal cord injury: a comprehensive review and practical recommendations. Br J Nutr 2022; 128:863-887. [PMID: 34551839 PMCID: PMC9389429 DOI: 10.1017/s0007114521003822] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Many persons with spinal cord injury (SCI) have one or more preventable chronic diseases related to excessive energetic intake and poor eating patterns. Appropriate nutrient consumption relative to need becomes a concern despite authoritative dietary recommendations from around the world. These recommendations were developed for the non-disabled population and do not account for the injury-induced changes in body composition, hypometabolic rate, hormonal dysregulation and nutrition status after SCI. Because evidence-based dietary reference intake values for SCI do not exist, ensuring appropriate consumption of macronutrient and micronutrients for their energy requirements becomes a challenge. In this compressive review, we briefly evaluate aspects of energy balance and appetite control relative to SCI. We report on the evidence regarding energy expenditure, nutrient intake and their relationship after SCI. We compare these data with several established nutritional guidelines from American Heart Association, Australian Dietary Guidelines, Dietary Guidelines for Americans, Institute of Medicine Dietary Reference Intake, Public Health England Government Dietary Recommendations, WHO Healthy Diet and the Paralyzed Veterans of America (PVA) Clinical Practice Guidelines. We also provide practical assessment and nutritional recommendations to facilitate a healthy dietary pattern after SCI. Because of a lack of strong SCI research, there are currently limited dietary recommendations outside of the PVA guidelines that capture the unique nutrient needs after SCI. Future multicentre clinical trials are needed to develop comprehensive, evidence-based dietary reference values specific for persons with SCI across the care continuum that rely on accurate, individual assessment of energy need.
Collapse
Affiliation(s)
- Gary J. Farkas
- Department of Physical Medicine and Rehabilitation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alicia Sneij
- Department of Physical Medicine and Rehabilitation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - David W. McMillan
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Eduard Tiozzo
- Department of Physical Medicine and Rehabilitation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mark S. Nash
- Department of Physical Medicine and Rehabilitation, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
- South Florida Spinal Cord Injury Model System, University of Miami Miller School of Medicine, Miami, FL, USA
| | - David R. Gater
- Department of Physical Medicine and Rehabilitation, University of Miami Miller School of Medicine, Miami, FL, USA
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
- South Florida Spinal Cord Injury Model System, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
29
|
Hoffman S, Alvares D, Adeli K. GLP-1 attenuates intestinal fat absorption and chylomicron production via vagal afferent nerves originating in the portal vein. Mol Metab 2022; 65:101590. [PMID: 36067913 PMCID: PMC9486018 DOI: 10.1016/j.molmet.2022.101590] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/22/2022] [Accepted: 09/01/2022] [Indexed: 12/04/2022] Open
Abstract
Background/Objective GLP-1R agonists have been shown to reduce fasting and postprandial plasma lipids, both of which are independent risk factors for the development of cardiovascular disease. However, how endogenous GLP-1 – which is rapidly degraded – modulates intestinal and hepatic lipid metabolism is less clear. A vagal gut-brain-axis originating in the portal vein has been proposed as a possible mechanism for GLP-1’s anti-lipemic effects. Here we sought to examine the relationship between vagal GLP-1 signalling and intestinal lipid absorption and lipoprotein production. Methods Syrian golden hamsters or C57BL/6 mice received portal vein injections of GLP-1(7-36), and postprandial and fasting plasma TG, TRL TG, or VLDL TG were examined. These experiments were repeated during sympathetic blockade, and under a variety of pharmacological or surgical deafferentation techniques. In addition, hamsters received nodose ganglia injections of a GLP-1R agonist or antagonist to further probe the vagal pathway. Peripheral studies were repeated in a novel GLP-1R KO hamster model and in our diet-induced hamster models of insulin resistance. Results GLP-1(7-36) site-specifically reduced postprandial and fasting plasma lipids in both hamsters and mice. These inhibitory effects of GLP-1 were investigated via pharmacological and surgical denervation experiments and found to be dependent on intact afferent vagal signalling cascades and efferent changes in sympathetic tone. Furthermore, GLP-1R agonism in the nodose ganglia resulted in markedly reduced postprandial plasma TG and TRL TG, and fasting VLDL TG and this nodose GLP-1R activity was essential for portal GLP-1s effect. Notably, portal and nodose ganglia GLP-1 effects were lost in GLP-1R KO hamsters and following diet-induced insulin resistance. Conclusion Our data demonstrates for the first time that portal GLP-1 modulates postprandial and fasting lipids via a complex vagal gut–brain–liver axis. Importantly, loss or interference with this signalling axis via surgical, pharmacological, or dietary intervention resulted in the loss of portal GLP-1s anti-lipemic effects. This supports emerging evidence that native GLP-1 works primarily through a vagal neuroendocrine mechanism.
Collapse
Affiliation(s)
- Simon Hoffman
- Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.
| | - Danielle Alvares
- Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.
| | - Khosrow Adeli
- Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.
| |
Collapse
|
30
|
Mikhailova EV, Derkach KV, Shpakov AO, Romanova IV. Melanocortin 1 Receptors in the Hypothalamus of Mice within the Norm and in Diet-Induced Obesity. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022040263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
Coker CR, White M, Singal A, Bingaman SS, Paul A, Arnold AC, Silberman Y. Minocycline Reduces Hypothalamic Microglia Activation and Improves Metabolic Dysfunction in High Fat Diet-Induced Obese Mice. Front Physiol 2022; 13:933706. [PMID: 35784876 PMCID: PMC9244633 DOI: 10.3389/fphys.2022.933706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 05/27/2022] [Indexed: 01/27/2023] Open
Abstract
Obesity is associated with insulin resistance, glucose intolerance, inflammation, and altered neuronal activity in brain regions controlling metabolic functions including food intake, energy expenditure, and glucose homeostasis, such as the hypothalamus. In this study, we tested the hypothesis that inhibiting inflammation with minocycline could reduce adverse metabolic consequences associated with high-fat diet (HFD)-induced obesity in mice and sought to determine if metabolic improvements were associated with reduced hypothalamic microglia activity. Male C57Bl/6J mice were placed on 60% HFD for 12 weeks, with minocycline (40 mg/kg, p.o.) or normal tap water given during the last 6 weeks of diet. Age-matched mice maintained on control diet were used as an additional comparator group. Metabolic function was assessed during the last week of treatment. Ramified (resting) and non-ramified (active) microglia were quantified in the hypothalamus following immunohistochemical staining of ionized calcium-binding adaptor 1 (Iba-1) and further assessed by RNAseq. In HFD fed mice, minocycline attenuated body mass and adiposity without altering food intake suggesting enhanced energy expenditure. Minocycline also attenuated hyperinsulinemia and improved insulin sensitivity in HFD mice. Increased microglial activation and autophagy gene network changes were observed in the paraventricular nucleus (PVN) of the hypothalamus of HFD mice, which was prevented by minocycline treatment. Contrary to PVN findings, there were no significant effects of either HFD or minocycline on microglia activation in the hypothalamic arcuate nucleus or central amygdala. Together, these findings suggest that minocycline improves HFD-induced weight gain and insulin resistance in part by reducing inflammatory processes in the PVN, a key hypothalamic region regulating metabolic function.
Collapse
Affiliation(s)
- Caitlin R. Coker
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, United States
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University School of Medicine, Washington, DC, United States
| | - Melissa White
- Department of Comparative Medicine, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Aneesh Singal
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Sarah S. Bingaman
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Anirban Paul
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Amy C. Arnold
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Yuval Silberman
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| |
Collapse
|
32
|
Casagrande BP, Bueno AA, Pisani LP, Estadella D. Hepatic glycogen participates in the regulation of hypothalamic pAkt/Akt ratio in high-sugar/high-fat diet-induced obesity. Metab Brain Dis 2022; 37:1423-1434. [PMID: 35316448 DOI: 10.1007/s11011-022-00944-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/22/2022] [Indexed: 01/07/2023]
Abstract
The hypothalamus is a major integrating centre that controls energy homeostasis and plays a major role in hepatic glycogen (HGlyc) turnover. Not only do hypothalamic and hepatic Akt levels influence glucose homeostasis and glycogen synthesis, but exposure to high-sugar/high-fat diets (HSHF) can also lead to hypothalamic inflammation and HGlyc accumulation. HSHF withdrawal overall restores energy and glucose homeostasis, but the actual relationship between hypothalamic inflammation and HGlyc after short-term HSHF withdrawal has not yet been fully elucidated. Here we investigated the short-term effects of HSHF withdrawal preceded by a 30-day HSHF intake on the liver-hypothalamus crosstalk and glucose homeostasis. Sixty-day old male Wistar rats were fed for 30 days a control chow (n = 10) (Ct), or an HSHF diet (n = 20). On the 30th day of dietary intervention, a random HSHF subset (n = 10) had their diets switched to control chow for 48 h (Hw) whilst the remaining HSHF rats remained in the HSHF diet (n = 10) (Hd). All rats were anaesthetized and euthanized at the end of the protocol. We quantified HGlyc, Akt phosphorylation, inflammation and glucose homeostasis biomarkers. We also assessed the effect of propensity to obesity on those biomarkers, as detailed previously. Hd rats showed impaired glucose homeostasis, higher HGlyc and hypothalamic inflammation, and lower pAkt/Akt. Increased HGlyc was significantly associated with HSHF intake on pAkt/Akt lowered levels. We also found that HGlyc breakdown may have prevented a further pAkt/Akt drop after HSHF withdrawal. Propensity to obesity showed no apparent effect on hypothalamic inflammation or glucose homeostasis. Our findings suggest a comprehensive role of HGlyc as a structural and functional modulator of energy metabolism, and such roles may come into play relatively rapidly.
Collapse
Affiliation(s)
- Breno P Casagrande
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo-UNIFESP/BS, 1015-020, Santos, São Paulo, Brazil
| | - Allain A Bueno
- College of Health, Life and Environmental Sciences, University of Worcester, Henwick Grove, WR2 6AJ, Worcester, United Kingdom
| | - Luciana P Pisani
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo-UNIFESP/BS, 1015-020, Santos, São Paulo, Brazil
| | - Debora Estadella
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo-UNIFESP/BS, 1015-020, Santos, São Paulo, Brazil.
| |
Collapse
|
33
|
Rapuano KM, Berrian N, Baskin-Sommers A, Décarie-Spain L, Sharma S, Fulton S, Casey BJ, Watts R. Longitudinal Evidence of a Vicious Cycle Between Nucleus Accumbens Microstructure and Childhood Weight Gain. J Adolesc Health 2022; 70:961-969. [PMID: 35248457 PMCID: PMC9133207 DOI: 10.1016/j.jadohealth.2022.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 12/27/2021] [Accepted: 01/04/2022] [Indexed: 10/19/2022]
Abstract
PURPOSE Pediatric obesity is a growing public health concern. Previous work has observed diet to impact nucleus accumbens (NAcc) inflammation in rodents, measured by the reactive proliferation of glial cells. Recent work in humans has demonstrated a relationship between NAcc cell density-a proxy for neuroinflammation-and weight gain in youth; however, the directionality of this relationship in the developing brain and association with diet remains unknown. METHODS Waist circumference (WC) and NAcc cell density were collected in a large cohort of children (n > 2,000) participating in the Adolescent Brain Cognitive Development (ABCD) Study (release 3.0) at baseline (9-10 y) and at a Year 2 follow-up (11-12 y). Latent change score modeling (LCSM) was used to disentangle contributions of baseline measures to two-year changes in WC percentile and NAcc cellularity. In addition, the role of NAcc cellularity in mediating the relationship between diet and WC percentile was assessed using dietary intake data collected at Year 2. RESULTS LCSM indicates that baseline WC percentile influences change in NAcc cellularity and that baseline NAcc cell density influences change in WC percentile. NAcc cellularity was significantly associated with WC percentile at Year 2 and mediated the relationship between dietary fat consumption and WC percentile. CONCLUSIONS These results implicate a vicious cycle whereby NAcc cell density biases longitudinal changes in WC percentile and vice versa. Moreover, NAcc cell density may mediate the relationship between diet and weight gain in youth. These findings suggest that diet-induced inflammation of reward circuitry may lead to behavioral changes that further contribute to weight gain.
Collapse
Affiliation(s)
| | | | | | - Léa Décarie-Spain
- Department of Biological Sciences, University of Southern California
| | - Sandeep Sharma
- Department of Comparative Biology and Experimental Medicine, University of Calgary
| | - Stephanie Fulton
- Department of Nutrition, University of Montreal & Centre de Recherche du CHUM
| | - BJ Casey
- Department of Psychology, Yale University
| | | |
Collapse
|
34
|
The central nervous system control of energy homeostasis: high fat diet induced hypothalamic microinflammation and obesity. Brain Res Bull 2022; 185:99-106. [PMID: 35525336 DOI: 10.1016/j.brainresbull.2022.04.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/27/2022] [Accepted: 04/30/2022] [Indexed: 11/22/2022]
Abstract
Obesity is believed to arise through the imbalance of energy homeostasis controlled by the central nervous system, where the hypothalamus plays the fundamental role in energy metabolism. In this review, we will provide an overview regarding the functions of POMC neurons and AgRP neurons in acute nucleus of the hypothalamus which mediated the energy metabolism, highlighting their interactions with peripheral organs derived hormones in control of energy homeostasis. Furthermore, the role of high fat diet induced hypothalamic microinflammation in the pathogenesis of obesity will be discussed. We hope this review could help researchers to understand the mechanism of hypothalamus in control of energy metabolism, and design related drugs to block the pathways involving in the impaired metabolism in obese patients.
Collapse
|
35
|
Lama A, Pirozzi C, Severi I, Morgese MG, Senzacqua M, Annunziata C, Comella F, Del Piano F, Schiavone S, Petrosino S, Mollica MP, Diano S, Trabace L, Calignano A, Giordano A, Mattace Raso G, Meli R. Palmitoylethanolamide dampens neuroinflammation and anxiety-like behavior in obese mice. Brain Behav Immun 2022; 102:110-123. [PMID: 35176443 PMCID: PMC10662208 DOI: 10.1016/j.bbi.2022.02.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 02/01/2022] [Accepted: 02/04/2022] [Indexed: 12/11/2022] Open
Abstract
High-fat diet (HFD) consumption leads to obesity and a chronic state of low-grade inflammation, named metainflammation. Notably, metainflammation contributes to neuroinflammation due to the increased levels of circulating free fatty acids and cytokines. It indicates a strict interplay between peripheral and central counterparts in the pathogenic mechanisms of obesity-related mood disorders. In this context, the impairment of internal hypothalamic circuitry runs in tandem with the alteration of other brain areas associated with emotional processing (i.e., hippocampus and amygdala). Palmitoylethanolamide (PEA), an endogenous lipid mediator belonging to the N-acylethanolamines family, has been extensively studied for its pleiotropic effects both at central and peripheral level. Our study aimed to elucidate PEA capability in limiting obesity-induced anxiety-like behavior and neuroinflammation-related features in an experimental model of HFD-fed obese mice. PEA treatment promoted an improvement in anxiety-like behavior of obese mice and the systemic inflammation, reducing serum pro-inflammatory mediators (i.e., TNF-α, IL-1β, MCP-1, LPS). In the amygdala, PEA increased dopamine turnover, as well as GABA levels. PEA also counteracted the overactivation of HPA axis, reducing the expression of hypothalamic corticotropin-releasing hormone and its type 1 receptor. Moreover, PEA attenuated the immunoreactivity of Iba-1 and GFAP and reduced pro-inflammatory pathways and cytokine production in both the hypothalamus and hippocampus. This finding, together with the reduced transcription of mast cell markers (chymase 1 and tryptase β2) in the hippocampus, indicated the weakening of immune cell activation underlying the neuroprotective effect of PEA. Obesity-driven neuroinflammation was also associated with the disruption of blood-brain barrier (BBB) in the hippocampus. PEA limited the albumin extravasation and restored tight junction transcription modified by HFD. To gain mechanistic insight, we designed an in vitro model of metabolic injury using human neuroblastoma SH-SY5Y cells insulted by a mix of glucosamine and glucose. Here, PEA directly counteracted inflammation and mitochondrial dysfunction in a PPAR-α-dependent manner since the pharmacological blockade of the receptor reverted its effects. Our results strengthen the therapeutic potential of PEA in obesity-related neuropsychiatric comorbidities, controlling neuroinflammation, BBB disruption, and neurotransmitter imbalance involved in behavioral dysfunctions.
Collapse
Affiliation(s)
- Adriano Lama
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 49 - 80131 Naples, Italy
| | - Claudio Pirozzi
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 49 - 80131 Naples, Italy
| | - Ilenia Severi
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto, 10, A - 60020 Ancona, Italy
| | - Maria Grazia Morgese
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20 - 71122 Foggia, Italy
| | - Martina Senzacqua
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto, 10, A - 60020 Ancona, Italy
| | - Chiara Annunziata
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 49 - 80131 Naples, Italy
| | - Federica Comella
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 49 - 80131 Naples, Italy
| | - Filomena Del Piano
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Via Delpino 1, 80137 Naples, Italy
| | - Stefania Schiavone
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20 - 71122 Foggia, Italy
| | - Stefania Petrosino
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Napoli, Italy
| | - Maria Pina Mollica
- Department of Biology, University of Naples Federico II, Complesso Universitario di Monte, Sant'Angelo, Cupa Nuova Cinthia 21 - Edificio 7, 80126 Naples, Italy
| | - Sabrina Diano
- Program in Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20 - 71122 Foggia, Italy
| | - Antonio Calignano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 49 - 80131 Naples, Italy
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto, 10, A - 60020 Ancona, Italy
| | - Giuseppina Mattace Raso
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 49 - 80131 Naples, Italy.
| | - Rosaria Meli
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 49 - 80131 Naples, Italy
| |
Collapse
|
36
|
Folick A, Cheang RT, Valdearcos M, Koliwad SK. Metabolic factors in the regulation of hypothalamic innate immune responses in obesity. Exp Mol Med 2022; 54:393-402. [PMID: 35474339 PMCID: PMC9076660 DOI: 10.1038/s12276-021-00666-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 05/13/2021] [Indexed: 12/14/2022] Open
Abstract
The hypothalamus is a central regulator of body weight and energy homeostasis. There is increasing evidence that innate immune activation in the mediobasal hypothalamus (MBH) is a key element in the pathogenesis of diet-induced obesity. Microglia, the resident immune cells in the brain parenchyma, have been shown to play roles in diverse aspects of brain function, including circuit refinement and synaptic pruning. As such, microglia have also been implicated in the development and progression of neurological diseases. Microglia express receptors for and are responsive to a wide variety of nutritional, hormonal, and immunological signals that modulate their distinct functions across different brain regions. We showed that microglia within the MBH sense and respond to a high-fat diet and regulate the function of hypothalamic neurons to promote food intake and obesity. Neurons, glia, and immune cells within the MBH are positioned to sense and respond to circulating signals that regulate their capacity to coordinate aspects of systemic energy metabolism. Here, we review the current knowledge of how these peripheral signals modulate the innate immune response in the MBH and enable microglia to regulate metabolic control.
Collapse
Affiliation(s)
- Andrew Folick
- Diabetes Center and Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA
| | - Rachel T Cheang
- Diabetes Center and Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA
| | - Martin Valdearcos
- Diabetes Center and Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA.
| | - Suneil K Koliwad
- Diabetes Center and Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA.
| |
Collapse
|
37
|
Eroğlu FE, Sanlier N. Effect of fermented foods on some neurological diseases, microbiota, behaviors: mini review. Crit Rev Food Sci Nutr 2022; 63:8066-8082. [PMID: 35317694 DOI: 10.1080/10408398.2022.2053060] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Fermented foods are among the traditional foods consumed for centuries. In recent years, awareness of fermented foods has been increasing due to their positive health benefits. Fermented foods contain beneficial microorganisms. Fermented foods, such as kefir, kimchi, sauerkraut, and yoghurt, contain Lactic acid bacteria (LAB), such as Lactobacilli, Bifidobacteria, and their primary metabolites (lactic acid). Although studies on the effect of consumption of fermented foods on diabetes, cardiovascular, obesity, gastrointestinal diseases on chronic diseases have been conducted, more studies are needed regarding the relationship between neurological diseases and microbiota. There are still unexplored mechanisms in the relationship between the brain and intestine. In this review, we answer how the consumption of fermented foods affects the brain and behavior of Alzheimer's disease, Parkinson's disease, multiple sclerosis disease, stroke, and gut microbiota.
Collapse
Affiliation(s)
- Fatma Elif Eroğlu
- Department of Nutrition and Dietetics, Ankara Medipol University, Institute of Health Sciences, Ankara, Turkey
| | - Nevin Sanlier
- Department of Nutrition and Dietetics, School of Health Sciences, Ankara Medipol University, Altındağ, Ankara, Turkey
| |
Collapse
|
38
|
Lyu Y, Liu D, Nguyen P, Peters I, Heilmann RM, Fievez V, Hemeryck LY, Hesta M. Differences in Metabolic Profiles of Healthy Dogs Fed a High-Fat vs. a High-Starch Diet. Front Vet Sci 2022; 9:801863. [PMID: 35252418 PMCID: PMC8891928 DOI: 10.3389/fvets.2022.801863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/24/2022] [Indexed: 11/17/2022] Open
Abstract
Obesity is a common problem in dogs and overconsumption of energy-rich foods is a key factor. This study compared the inflammatory response and fecal metabolome of dogs fed a high-fat vs. a high-starch diet. Ten healthy lean adult beagles were equally allocated into two groups in a cross-over design. Each group received two diets in which fat (horse fat) and starch (pregelatinized corn starch) were exchanged in an isocaloric way to compare high fat vs. high starch. There was a tendency to increase the glucose and glycine concentrations and the glucose/insulin ratio in the blood in dogs fed with the high-fat diet, whereas there was a decrease in the level of Non-esterified fatty acids and a tendency to decrease the alanine level in dogs fed with the high-starch diet. Untargeted analysis of the fecal metabolome revealed 10 annotated metabolites of interest, including L-methionine, which showed a higher abundance in dogs fed the high-starch diet. Five other metabolites were upregulated in dogs fed the high-fat diet, but could not be annotated. The obtained results indicate that a high-starch diet, compared to a high-fat diet, may promote lipid metabolism, anti-oxidative effects, protein biosynthesis and catabolism, mucosal barrier function, and immunomodulation in healthy lean dogs.
Collapse
Affiliation(s)
- Yang Lyu
- ECAN Equine and Companion Animal Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Daisy Liu
- ECAN Equine and Companion Animal Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Patrick Nguyen
- Nutrition, Physiopathology and Pharmacology Unit, National College of Veterinary Medicine, Food Science and Engineering, Nantes, France
| | - Iain Peters
- SYNLAB VPG, Exeter Science Park, Exeter, United Kingdom
| | - Romy M. Heilmann
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Veerle Fievez
- Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent, Belgium
| | - Lieselot Y. Hemeryck
- Laboratory of Chemical Analysis, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- *Correspondence: Myriam Hesta
| | - Myriam Hesta
- ECAN Equine and Companion Animal Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Lieselot Y. Hemeryck
| |
Collapse
|
39
|
Song X, Wang L, Liu Y, Zhang X, Weng P, Liu L, Zhang R, Wu Z. The gut microbiota–brain axis: Role of the gut microbial metabolites of dietary food in obesity. Food Res Int 2022; 153:110971. [DOI: 10.1016/j.foodres.2022.110971] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 01/27/2022] [Accepted: 01/30/2022] [Indexed: 12/13/2022]
|
40
|
Depression and obesity among females, are sex specificities considered? Arch Womens Ment Health 2021; 24:851-866. [PMID: 33880649 DOI: 10.1007/s00737-021-01123-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 03/21/2021] [Indexed: 12/13/2022]
Abstract
This study aimed to systematically review the relationship of obesity-depression in the female sex. We carried out a systematic search (PubMed, MEDLINE, Embase) to quantify the articles (controlled trials and randomized controlled trials) regarding obesity and depression on a female population or a mixed sample. Successively, we established whether the sex specificities were studied by the authors and if they reported on collecting data regarding factors that may contribute to the evolution of obesity and depression and that could be responsible for the greater susceptibility of females to those conditions. After applying the inclusion and exclusion criteria, we found a total of 20 articles with a female sample and 54 articles with a mixed sample. More than half of all articles (51.35%, n = 38) evaluated the relationship between depression and obesity, but only 20 (27.03%) evaluated this relationship among females; still, 80% of those (n = 16) presented supporting results. However, few articles considered confounding factors related to female hormones (12.16%, n = 9) and none of the articles focused on factors responsible for the binomial obesity-depression in the female sex. The resulting articles also supported that depression (and related impairments) influencing obesity (and related impairments) is a two-way road. This systematic review supports the concurrency of obesity-depression in females but also shows how sex specificities are ultimately under-investigated. Female sex specificity is not being actively considered when studying the binomial obesity-depression, even within a female sample. Future studies should focus on trying to understand how the female sex and normal hormonal variations influence these conditions.
Collapse
|
41
|
Long-term diet-induced obesity does not lead to learning and memory impairment in adult mice. PLoS One 2021; 16:e0257921. [PMID: 34587222 PMCID: PMC8480843 DOI: 10.1371/journal.pone.0257921] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 09/13/2021] [Indexed: 02/06/2023] Open
Abstract
Obesity arising from excessive dietary fat intake is a risk factor for cognitive decline, dementia and neurodegenerative diseases, including Alzheimer’s disease. Here, we studied the effect of long-term high-fat diet (HFD) (24 weeks) and return to normal diet (ND) on behavioral features, microglia and neurons in adult male C57BL/6J mice. Consequences of HFD-induced obesity and dietary changes on general health (coat appearance, presence of vibrissae), sensory and motor reflexes, learning and memory were assessed by applying a phenotypic assessment protocol, the Y maze and Morris Water Maze test. Neurons and microglia were histologically analyzed within the mediobasal hypothalamus, hippocampus and frontal motor cortex after long-term HFD and change of diet. Long periods of HFD caused general health issues (coat alterations, loss of vibrissae), but did not affect sensory and motor reflexes, emotional state, memory and learning. Long-term HFD increased the microglial response (increased Iba1 fluorescence intensity, percentage of Iba1-stained area and Iba1 gene expression) within the hypothalamus, but not in the cortex and hippocampus. In neither of these regions, neurodegeneration or intracellular lipid droplet accumulation was observed. The former alterations were reversible in mice whose diet was changed from HFD to ND. Taken together, long periods of excessive dietary fat alone do not cause learning deficits or spatial memory impairment, though HFD-induced obesity may have detrimental consequences for cognitive flexibility. Our data confirm the selective responsiveness of hypothalamic microglia to HFD.
Collapse
|
42
|
Qiu L, Chen M, Wang X, Chen S, Ying Z. PM2.5 Exposure of Mice during Spermatogenesis: A Role of Inhibitor κB Kinase 2 in Pro-Opiomelanocortin Neurons. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:97006. [PMID: 34495743 PMCID: PMC8425520 DOI: 10.1289/ehp8868] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 08/09/2021] [Accepted: 08/19/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Epidemiological studies have shown that exposure to ambient fine particulate matter with aerodynamic diameter less than or equal to 2.5 μm (PM2.5) correlates with a decrease in sperm count, but the biological mechanism remains elusive. OBJECTIVES This study tested whether hypothalamic inflammation, an emerging pathophysiological mediator, mediates the development of lower epididymal sperm count due to PM2.5 exposure. METHODS Inhibitor κB kinase 2 (IKK2) was conditionally knocked out either in all neurons or subtypes of hypothalamic neurons of mice. Effects of concentrated ambient PM2.5 (CAP) exposure on hypothalamic inflammation, the hypothalamic-pituitary-gonadal (HPG) axis, and epididymal sperm count of these mouse models were then assessed. Furthermore, to test whether hypothalamic inflammation is sufficient to decrease sperm production, we overexpressed constitutively active IKK2 (IKK2ca) either in all neurons or subtypes of hypothalamic neurons and assessed hypothalamic inflammation, the HPG axis, and sperm production of these overexpression mouse models. RESULTS CAP-exposed wild-type control mice vs. filtered air (FA)-exposed wild-type control mice had a higher expression of hypothalamic inflammatory markers, lower functional indexes of the HPG axis, and a lower epididymal sperm count. In contrast, all these measurements for CAP- vs. FA-exposed mice deficient of IKK2 in all neurons were comparable. We also found that overexpression of IKK2ca in either all neurons or pro-opiomelanocortin (POMC) neurons only, but not in Agouti-related protein (AgRP) neurons only, resulted in lower functional indexes of the HPG axis and a lower epididymal sperm count. Moreover, we showed that CAP- vs. FA-exposed mice deficient of IKK2 in POMC neurons had a comparable expression of hypothalamic inflammatory markers, comparable functional indexes of the HPG axis, and a comparable epididymal sperm count. DISCUSSION This mouse model study shows a causal role of IKK2 of POMC neurons in the development of lower epididymal sperm count due to PM2.5 exposure, providing a mechanistic insight into this emerging pathogenesis. https://doi.org/10.1289/EHP8868.
Collapse
Affiliation(s)
- Lianglin Qiu
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, Maryland, USA
- School of Public Health, Nantong University, Nantong, Jiangsu, China
| | - Minjie Chen
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Xiaoke Wang
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, Maryland, USA
- School of Public Health, Nantong University, Nantong, Jiangsu, China
| | - Sufang Chen
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, Maryland, USA
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhekang Ying
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
43
|
Rautmann AW, de La Serre CB. Microbiota's Role in Diet-Driven Alterations in Food Intake: Satiety, Energy Balance, and Reward. Nutrients 2021; 13:nu13093067. [PMID: 34578945 PMCID: PMC8470213 DOI: 10.3390/nu13093067] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/18/2021] [Accepted: 08/25/2021] [Indexed: 02/07/2023] Open
Abstract
The gut microbiota plays a key role in modulating host physiology and behavior, particularly feeding behavior and energy homeostasis. There is accumulating evidence demonstrating a role for gut microbiota in the etiology of obesity. In human and rodent studies, obesity and high-energy feeding are most consistently found to be associated with decreased bacterial diversity, changes in main phyla relative abundances and increased presence of pro-inflammatory products. Diet-associated alterations in microbiome composition are linked with weight gain, adiposity, and changes in ingestive behavior. There are multiple pathways through which the microbiome influences food intake. This review discusses these pathways, including peripheral mechanisms such as the regulation of gut satiety peptide release and alterations in leptin and cholecystokinin signaling along the vagus nerve, as well as central mechanisms, such as the modulation of hypothalamic neuroinflammation and alterations in reward signaling. Most research currently focuses on determining the role of the microbiome in the development of obesity and using microbiome manipulation to prevent diet-induced increase in food intake. More studies are necessary to determine whether microbiome manipulation after prolonged energy-dense diet exposure and obesity can reduce intake and promote meaningful weight loss.
Collapse
|
44
|
Berding K, Vlckova K, Marx W, Schellekens H, Stanton C, Clarke G, Jacka F, Dinan TG, Cryan JF. Diet and the Microbiota-Gut-Brain Axis: Sowing the Seeds of Good Mental Health. Adv Nutr 2021; 12:1239-1285. [PMID: 33693453 PMCID: PMC8321864 DOI: 10.1093/advances/nmaa181] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
Over the past decade, the gut microbiota has emerged as a key component in regulating brain processes and behavior. Diet is one of the major factors involved in shaping the gut microbiota composition across the lifespan. However, whether and how diet can affect the brain via its effects on the microbiota is only now beginning to receive attention. Several mechanisms for gut-to-brain communication have been identified, including microbial metabolites, immune, neuronal, and metabolic pathways, some of which could be prone to dietary modulation. Animal studies investigating the potential of nutritional interventions on the microbiota-gut-brain axis have led to advancements in our understanding of the role of diet in this bidirectional communication. In this review, we summarize the current state of the literature triangulating diet, microbiota, and host behavior/brain processes and discuss potential underlying mechanisms. Additionally, determinants of the responsiveness to a dietary intervention and evidence for the microbiota as an underlying modulator of the effect of diet on brain health are outlined. In particular, we emphasize the understudied use of whole-dietary approaches in this endeavor and the need for greater evidence from clinical populations. While promising results are reported, additional data, specifically from clinical cohorts, are required to provide evidence-based recommendations for the development of microbiota-targeted, whole-dietary strategies to improve brain and mental health.
Collapse
Affiliation(s)
| | | | - Wolfgang Marx
- Deakin University, iMPACT – the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, VIC,Australia
| | - Harriet Schellekens
- APC Microbiome Ireland, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Sciences, University College Cork, Cork, Ireland
| | - Felice Jacka
- Deakin University, iMPACT – the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, VIC,Australia
- Centre for Adolescent Health, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Black Dog Institute, Randwick, NSW, Australia
- College of Public Health, Medical & Veterinary Sciences, James Cook University, Douglas, QLD, Australia
| | - Timothy G Dinan
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Sciences, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
45
|
Ye L, Jia G, Li Y, Wang Y, Chen H, Yu L, Wu D. C1q/TNF-related protein 4 restores leptin sensitivity by downregulating NF-κB signaling and microglial activation. J Neuroinflammation 2021; 18:159. [PMID: 34275474 PMCID: PMC8286609 DOI: 10.1186/s12974-021-02167-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/05/2021] [Indexed: 12/29/2022] Open
Abstract
Objective C1qTNF-related protein 4 (CTRP4) acts in the hypothalamus to modulate food intake in diet-induced obese mice and has been shown to exert an anti-inflammatory effect on macrophages. Since high-fat diet-induced microglial activation and hypothalamic inflammation impair leptin signaling and increase food intake, we aimed to explore the potential connection between the anorexigenic effect of CTRP4 and the suppression of hypothalamic inflammation in mice with DIO. Methods Using an adenovirus-mediated hypothalamic CTRP4 overexpression model, we investigated the impact of CTRP4 on food intake and the hypothalamic leptin signaling pathway in diet-induced obese mice. Furthermore, central and plasma proinflammatory cytokines, including TNF-α and IL-6, were measured by Western blotting and ELISA. Changes in the hypothalamic NF-κB signaling cascade and microglial activation were also examined in vivo. In addition, NF-κB signaling and proinflammatory factors were investigated in BV-2 cells after CTRP4 intervention. Results We found that food intake was decreased, while leptin signaling was significantly improved in mice with DIO after CTRP4 overexpression. Central and peripheral TNF-α and IL-6 levels were reduced by central Ad-CTRP4 administration. Hypothalamic NF-κB signaling and microglial activation were also significantly suppressed in vivo. In addition, NF-κB signaling was inhibited in BV-2 cells following CTRP4 intervention, which was consistent with the decreased production of TNF-α and IL-6. Conclusions Our data indicate that CTRP4 reverses leptin resistance by inhibiting NF-κB-dependent microglial activation and hypothalamic inflammation. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02167-2.
Collapse
Affiliation(s)
- Liu Ye
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Gongwei Jia
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Yuejie Li
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Ying Wang
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Hong Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lehua Yu
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Dandong Wu
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
46
|
Clyburn C, Travagli RA, Arnold AC, Browning KN. DMV extrasynaptic NMDA receptors regulate caloric intake in rats. JCI Insight 2021; 6:139785. [PMID: 33764905 PMCID: PMC8262316 DOI: 10.1172/jci.insight.139785] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 03/24/2021] [Indexed: 11/17/2022] Open
Abstract
Acute high-fat diet (aHFD) exposure induces a brief period of hyperphagia before caloric balance is restored. Previous studies have demonstrated that this period of regulation is associated with activation of synaptic N-methyl-D-aspartate (NMDA) receptors on dorsal motor nucleus of the vagus (DMV) neurons, which increases vagal control of gastric functions. Our aim was to test the hypothesis that activation of DMV synaptic NMDA receptors occurs subsequent to activation of extrasynaptic NMDA receptors. Sprague-Dawley rats were fed a control or high-fat diet for 3-5 days prior to experimentation. Whole-cell patch-clamp recordings from gastric-projecting DMV neurons; in vivo recordings of gastric motility, tone, compliance, and emptying; and food intake studies were used to assess the effects of NMDA receptor antagonism on caloric regulation. After aHFD exposure, inhibition of extrasynaptic NMDA receptors prevented the synaptic NMDA receptor-mediated increase in glutamatergic transmission to DMV neurons, as well as the increase in gastric tone and motility, while chronic extrasynaptic NMDA receptor inhibition attenuated the regulation of caloric intake. After aHFD exposure, the regulation of food intake involved synaptic NMDA receptor-mediated currents, which occurred in response to extrasynaptic NMDA receptor activation. Understanding these events may provide a mechanistic basis for hyperphagia and may identify novel therapeutic targets for the treatment of obesity.
Collapse
|
47
|
Clyburn C, Browning KN. Glutamatergic plasticity within neurocircuits of the dorsal vagal complex and the regulation of gastric functions. Am J Physiol Gastrointest Liver Physiol 2021; 320:G880-G887. [PMID: 33730858 PMCID: PMC8202199 DOI: 10.1152/ajpgi.00014.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The meticulous regulation of the gastrointestinal (GI) tract is required for the coordination of gastric motility and emptying, intestinal secretion, absorption, and transit as well as for the overarching management of food intake and energy homeostasis. Disruption of GI functions is associated with the development of severe GI disorders and the alteration of food intake and caloric balance. Functional GI disorders as well as the dysregulation of energy balance and food intake are frequently associated with, or result from, alterations in the central regulation of GI control. The faithful and rapid transmission of information from the stomach and upper GI tract to second-order neurons of the nucleus of the tractus solitarius (NTS) relies on the delicate modulation of excitatory glutamatergic transmission, as does the relay of integrated signals from the NTS to parasympathetic efferent neurons of the dorsal motor nucleus of the vagus (DMV). Many studies have focused on understanding the physiological and pathophysiological modulation of these glutamatergic synapses, although their role in the control and regulation of GI functions has lagged behind that of cardiovascular and respiratory functions. The purpose of this review is to examine the current literature exploring the role of glutamatergic transmission in the DVC in the regulation of GI functions.
Collapse
Affiliation(s)
- Courtney Clyburn
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, Pennsylvania
| | - Kirsteen N. Browning
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
48
|
Tran M, Wu J, Wang L, Shin DJ. A Potential Role for SerpinA3N in Acetaminophen-Induced Hepatotoxicity. Mol Pharmacol 2021; 99:277-285. [PMID: 33436521 PMCID: PMC7985612 DOI: 10.1124/molpharm.120.000117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/31/2020] [Indexed: 10/25/2022] Open
Abstract
Acetaminophen (APAP) is a commonly used pain and fever reliever but is also the most frequent cause of drug-induced liver injury. The mechanism pertaining acetaminophen toxicity has been well documented, whereas mechanisms of hepatotoxicity are not well established. Serine (or cysteine) peptidase inhibitor, clade A, member 3N (SerpinA3N), a serine protease inhibitor, is synthesized in the liver but the role of SerpinA3N in relation to APAP-induced liver injury is not known. Wild-type and hepatocyte-specific SerpinA3N knockout (HKO) mice were injected intraperitoneally with a single dose of PBS or APAP (400 mg/kg) for 12 hours, and markers of liver injury, cell death, and inflammation were assessed. SerpinA3N expression was highly induced in mice with APAP overdose. SerpinA3N HKO mice had diminished liver injury and necrosis as shown by lower alanine aminotransferase and interleukin-6 levels, accompanied by suppressed inflammatory cytokines and reduced neutrophil infiltration. The reduced oxidative stress was associated with enhanced antioxidant enzyme capabilities. Taken together, hepatocyte SerpinA3N deficiency reduced APAP-induced liver injury by ameliorating inflammation and modulating the 5' AMP-activated protein kinase-unc-51-like autophagy activating kinase 1 signaling pathway. Our study provides novel insights into a potential role for SerpinA3N in APAP-induced liver injury. SIGNIFICANCE STATEMENT: Our studies indicate that serine (or cysteine) peptidase inhibitor, clade A, member 3N (SerpinA3N) may have a pathophysiological role in modulating acetaminophen (APAP)-induced liver injury. More specifically, mice with hepatic deletion of SerpinA3N suppressed inflammation and liver injury to reduce APAP-induced hepatotoxicity. Controlling the inflammatory response offers possible approaches for novel therapeutics; therefore, understanding the pathophysiological role of SerpinA3N in inducing liver injury may add to the development of more efficacious treatments.
Collapse
Affiliation(s)
- Melanie Tran
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut (M.T., J.W., D.-J.S.) and Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, Connecticut (L.W.)
| | - Jianguo Wu
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut (M.T., J.W., D.-J.S.) and Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, Connecticut (L.W.)
| | - Li Wang
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut (M.T., J.W., D.-J.S.) and Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, Connecticut (L.W.)
| | - Dong-Ju Shin
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut (M.T., J.W., D.-J.S.) and Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, Connecticut (L.W.)
| |
Collapse
|
49
|
Mapping of Microglial Brain Region, Sex and Age Heterogeneity in Obesity. Int J Mol Sci 2021; 22:ijms22063141. [PMID: 33808700 PMCID: PMC8003547 DOI: 10.3390/ijms22063141] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/27/2022] Open
Abstract
The prevalence of obesity has increased rapidly in recent years and has put a huge burden on healthcare worldwide. Obesity is associated with an increased risk for many comorbidities, such as cardiovascular diseases, type 2 diabetes and hypertension. The hypothalamus is a key brain region involved in the regulation of food intake and energy expenditure. Research on experimental animals has shown neuronal loss, as well as microglial activation in the hypothalamus, due to dietary-induced obesity. Microglia, the resident immune cells in the brain, are responsible for maintaining the brain homeostasis and, thus, providing an optimal environment for neuronal function. Interestingly, in obesity, microglial cells not only get activated in the hypothalamus but in other brain regions as well. Obesity is also highly associated with changes in hippocampal function, which could ultimately result in cognitive decline and dementia. Moreover, changes have also been reported in the striatum and cortex. Microglial heterogeneity is still poorly understood, not only in the context of brain region but, also, age and sex. This review will provide an overview of the currently available data on the phenotypic differences of microglial innate immunity in obesity, dependent on brain region, sex and age.
Collapse
|
50
|
Fouesnard M, Zoppi J, Petera M, Le Gleau L, Migné C, Devime F, Durand S, Benani A, Chaffron S, Douard V, Boudry G. Dietary switch to Western diet induces hypothalamic adaptation associated with gut microbiota dysbiosis in rats. Int J Obes (Lond) 2021; 45:1271-1283. [PMID: 33714973 DOI: 10.1038/s41366-021-00796-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 02/01/2021] [Accepted: 02/23/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Early hyperphagia and hypothalamic inflammation encountered after Western diet (WD) are linked to rodent propensity to obesity. Inflammation in several brain structures has been associated with gut dysbiosis. Since gut microbiota is highly sensitive to dietary changes, we hypothesised that immediate gut microbiota adaptation to WD in rats is involved in inflammation-related hypothalamic modifications. METHODS We evaluated short-term impact of WD consumption (2 h, 1, 2 and 4 days) on hypothalamic metabolome and caecal microbiota composition and metabolome. Data integration analyses were performed to uncover potential relationships among these three datasets. Finally, changes in hypothalamic gene expression in absence of gut microbiota were evaluated in germ-free rats fed WD for 2 days. RESULTS WD quickly and profoundly affected the levels of several hypothalamic metabolites, especially oxidative stress markers. In parallel, WD consumption reduced caecal microbiota diversity, modified its composition towards pro-inflammatory profile and changed caecal metabolome. Data integration identified strong correlations between gut microbiota sub-networks, unidentified caecal metabolites and hypothalamic oxidative stress metabolites. Germ-free rats displayed reduced energy intake and no changes in redox homoeostasis machinery expression or pro-inflammatory cytokines after 2 days of WD, in contrast to conventional rats, which exhibited increased SOD2, GLRX and IL-6 mRNA levels. CONCLUSION A potentially pro-inflammatory gut microbiota and an early hypothalamic oxidative stress appear shortly after WD introduction. Tripartite data integration highlighted putative links between gut microbiota sub-networks and hypothalamic oxidative stress. Together with the absence of hypothalamic modifications in germ-free rats, this strongly suggests the involvement of the microbiota-hypothalamus axis in rat adaptation to WD introduction and in energy homoeostasis regulation.
Collapse
Affiliation(s)
| | | | - Mélanie Petera
- Clermont Auvergne University, INRAE, UNH, Plateforme d'Exploration du Métabolisme, MetaboHUB Clermont, Clermont-Ferrand, France
| | - Léa Le Gleau
- Institut MICALIS, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Carole Migné
- Clermont Auvergne University, INRAE, UNH, Plateforme d'Exploration du Métabolisme, MetaboHUB Clermont, Clermont-Ferrand, France
| | - Fabienne Devime
- Institut MICALIS, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Stéphanie Durand
- Clermont Auvergne University, INRAE, UNH, Plateforme d'Exploration du Métabolisme, MetaboHUB Clermont, Clermont-Ferrand, France
| | - Alexandre Benani
- Centre des Sciences du Goût et de l'Alimentation, Unité Mixte de Recherche 6265-Centre National de la Recherche Scientifique 13241-Institut National de la Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Université de Bourgogne, Dijon, France
| | - Samuel Chaffron
- Université de Nantes, CNRS (UMR6004), LS2N, Nantes, France.,Research Federation (FR2022) Tara Oceans GO-SEE, Paris, France
| | - Véronique Douard
- Institut MICALIS, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Gaëlle Boudry
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France.
| |
Collapse
|