51
|
Espinoza L, Fedorchak S, Boychuk CR. Interplay Between Systemic Metabolic Cues and Autonomic Output: Connecting Cardiometabolic Function and Parasympathetic Circuits. Front Physiol 2021; 12:624595. [PMID: 33776789 PMCID: PMC7991741 DOI: 10.3389/fphys.2021.624595] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/12/2021] [Indexed: 12/21/2022] Open
Abstract
There is consensus that the heart is innervated by both the parasympathetic and sympathetic nervous system. However, the role of the parasympathetic nervous system in controlling cardiac function has received significantly less attention than the sympathetic nervous system. New neuromodulatory strategies have renewed interest in the potential of parasympathetic (or vagal) motor output to treat cardiovascular disease and poor cardiac function. This renewed interest emphasizes a critical need to better understand how vagal motor output is generated and regulated. With clear clinical links between cardiovascular and metabolic diseases, addressing this gap in knowledge is undeniably critical to our understanding of the interaction between metabolic cues and vagal motor output, notwithstanding the classical role of the parasympathetic nervous system in regulating gastrointestinal function and energy homeostasis. For this reason, this review focuses on the central, vagal circuits involved in sensing metabolic state(s) and enacting vagal motor output to influence cardiac function. It will review our current understanding of brainstem vagal circuits and their unique position to integrate metabolic signaling into cardiac activity. This will include an overview of not only how metabolic cues alter vagal brainstem circuits, but also how vagal motor output might influence overall systemic concentrations of metabolic cues known to act on the cardiac tissue. Overall, this review proposes that the vagal brainstem circuits provide an integrative network capable of regulating and responding to metabolic cues to control cardiac function.
Collapse
Affiliation(s)
- Liliana Espinoza
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Stephanie Fedorchak
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Carie R Boychuk
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
52
|
Browning KN, Carson KE. Central Neurocircuits Regulating Food Intake in Response to Gut Inputs-Preclinical Evidence. Nutrients 2021; 13:nu13030908. [PMID: 33799575 PMCID: PMC7998662 DOI: 10.3390/nu13030908] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/02/2021] [Accepted: 03/07/2021] [Indexed: 02/07/2023] Open
Abstract
The regulation of energy balance requires the complex integration of homeostatic and hedonic pathways, but sensory inputs from the gastrointestinal (GI) tract are increasingly recognized as playing critical roles. The stomach and small intestine relay sensory information to the central nervous system (CNS) via the sensory afferent vagus nerve. This vast volume of complex sensory information is received by neurons of the nucleus of the tractus solitarius (NTS) and is integrated with responses to circulating factors as well as descending inputs from the brainstem, midbrain, and forebrain nuclei involved in autonomic regulation. The integrated signal is relayed to the adjacent dorsal motor nucleus of the vagus (DMV), which supplies the motor output response via the efferent vagus nerve to regulate and modulate gastric motility, tone, secretion, and emptying, as well as intestinal motility and transit; the precise coordination of these responses is essential for the control of meal size, meal termination, and nutrient absorption. The interconnectivity of the NTS implies that many other CNS areas are capable of modulating vagal efferent output, emphasized by the many CNS disorders associated with dysregulated GI functions including feeding. This review will summarize the role of major CNS centers to gut-related inputs in the regulation of gastric function with specific reference to the regulation of food intake.
Collapse
|
53
|
Gao R, Fu Q, Jiang HM, Shen M, Zhao RL, Qian Y, He YQ, Xu KF, Xu XY, Chen H, Zhang Q, Yang T. Temporal metabolic and transcriptomic characteristics crossing islets and liver reveal dynamic pathophysiology in diet-induced diabetes. iScience 2021; 24:102265. [PMID: 33817571 PMCID: PMC8008187 DOI: 10.1016/j.isci.2021.102265] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/30/2020] [Accepted: 03/01/2021] [Indexed: 12/15/2022] Open
Abstract
To investigate the molecular mechanisms underlying islet dysfunction and insulin resistance in diet-induced diabetes, we conducted temporal RNA sequencing of tissues responsible for insulin secretion (islets) and action (liver) every 4 weeks in mice on high-fat (HFD) or chow diet for 24 weeks, linking to longitudinal profile of metabolic characteristics. The diverse responses of α, β, and δ cells to glucose and palmitate indicated HFD-induced dynamic deterioration of islet function from dysregulation to failure. Insulin resistance developed with variable time course in different tissues. Weighted gene co-expression network analysis and Ingenuity Pathway Analysis implicated islets and liver jointly programmed β-cell compensatory adaption via cell proliferation at early phase and irreversible islet dysfunction by inappropriate immune response at later stage, and identified interconnected molecules including growth differentiation factor 15. Frequencies of T cell subpopulation showed an early decrement in Tregs followed by increases in Th1 and Th17 cells during progression to diabetes.
Collapse
Affiliation(s)
- Rui Gao
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China.,Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX37LE, UK
| | - Qi Fu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - He-Min Jiang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Min Shen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Rui-Ling Zhao
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yu Qian
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yun-Qiang He
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Kuan-Feng Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xin-Yu Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Heng Chen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Quan Zhang
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX37LE, UK
| | - Tao Yang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
54
|
Neuroprotective Effects of Testosterone in the Hypothalamus of an Animal Model of Metabolic Syndrome. Int J Mol Sci 2021; 22:ijms22041589. [PMID: 33557413 PMCID: PMC7914611 DOI: 10.3390/ijms22041589] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/31/2021] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
Metabolic syndrome (MetS) is known to be associated to inflammation and alteration in the hypothalamus, a brain region implicated in the control of several physiological functions, including energy homeostasis and reproduction. Previous studies demonstrated the beneficial effects of testosterone treatment (TTh) in counteracting some MetS symptoms in both animal models and clinical studies. This study investigated the effect of TTh (30 mg/kg/week for 12 weeks) on the hypothalamus in a high-fat diet (HFD)-induced animal model of MetS, utilizing quantitative RT-PCR and immunohistochemical analyses. The animal model recapitulates the human MetS features, including low testosterone/gonadotropin plasma levels. TTh significantly improved MetS-induced hypertension, visceral adipose tissue accumulation, and glucose homeostasis derangements. Within hypothalamus, TTh significantly counteracted HFD-induced inflammation, as detected in terms of expression of inflammatory markers and microglial activation. Moreover, TTh remarkably reverted the HFD-associated alterations in the expression of important regulators of energy status and reproduction, such as the melanocortin and the GnRH-controlling network. Our results suggest that TTh may exert neuroprotective effects on the HFD-related hypothalamic alterations, with positive outcomes on the circuits implicated in the control of energy metabolism and reproductive tasks, thus supporting a possible role of TTh in the clinical management of MetS.
Collapse
|
55
|
Coker CR, Keller BN, Arnold AC, Silberman Y. Impact of High Fat Diet and Ethanol Consumption on Neurocircuitry Regulating Emotional Processing and Metabolic Function. Front Behav Neurosci 2021; 14:601111. [PMID: 33574742 PMCID: PMC7870708 DOI: 10.3389/fnbeh.2020.601111] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/28/2020] [Indexed: 01/12/2023] Open
Abstract
The prevalence of psychiatry disorders such as anxiety and depression has steadily increased in recent years in the United States. This increased risk for anxiety and depression is associated with excess weight gain, which is often due to over-consumption of western diets that are typically high in fat, as well as with binge eating disorders, which often overlap with overweight and obesity outcomes. This finding suggests that diet, particularly diets high in fat, may have important consequences on the neurocircuitry regulating emotional processing as well as metabolic functions. Depression and anxiety disorders are also often comorbid with alcohol and substance use disorders. It is well-characterized that many of the neurocircuits that become dysregulated by overconsumption of high fat foods are also involved in drug and alcohol use disorders, suggesting overlapping central dysfunction may be involved. Emerging preclinical data suggest that high fat diets may be an important contributor to increased susceptibility of binge drug and ethanol intake in animal models, suggesting diet could be an important aspect in the etiology of substance use disorders. Neuroinflammation in pivotal brain regions modulating metabolic function, food intake, and binge-like behaviors, such as the hypothalamus, mesolimbic dopamine circuits, and amygdala, may be a critical link between diet, ethanol, metabolic dysfunction, and neuropsychiatric conditions. This brief review will provide an overview of behavioral and physiological changes elicited by both diets high in fat and ethanol consumption, as well as some of their potential effects on neurocircuitry regulating emotional processing and metabolic function.
Collapse
Affiliation(s)
- Caitlin R. Coker
- Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC, United States
| | - Bailey N. Keller
- Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, United States
| | - Amy C. Arnold
- Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, United States
| | - Yuval Silberman
- Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, United States
| |
Collapse
|
56
|
Cawthon CR, Kirkland RA, Pandya S, Brinson NA, de La Serre CB. Non-neuronal crosstalk promotes an inflammatory response in nodose ganglia cultures after exposure to byproducts from gram positive, high-fat-diet-associated gut bacteria. Physiol Behav 2020; 226:113124. [PMID: 32763334 PMCID: PMC7530053 DOI: 10.1016/j.physbeh.2020.113124] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 02/06/2023]
Abstract
Vagal afferent neurons (VAN) projecting to the lamina propria of the digestive tract are the primary source of gut-originating signals to the central nervous system (CNS). VAN cell bodies are found in the nodose ganglia (NG). Responsiveness of VAN to gut-originating signals is altered by feeding status with sensitivity to satiety signals such as cholecystokinin (CCK) increasing in the fed state. Chronic high-fat (HF) feeding results in inflammation at the level of the NG associated with a loss of VAN ability to switch phenotype from the fasted to the fed state. HF feeding also leads to compositional changes in the gut microbiota. HF diet consumption notably drives increased Firmicutes to Bacteroidetes phyla ratio and increased members of the Actinobacteria phylum. Firmicutes and Actinobacteria are largely gram positive (GP). In this study, we aimed to determine if byproducts from GP bacteria can induce an inflammatory response in cultured NG and to characterize the mechanism and cell types involved in the response. NG were collected from male Wistar rats and cultured for a total of 72 hours. At 48-68 hours after plating, cultures were treated with neuronal culture media in which Serinicoccus chungangensis had been grown and removed (SUP), lipoteichoic acid (LTA), or meso-diaminopimelic acid (meso-DAP). Some treatments included the glial inhibitors minocycline (MINO) and/or fluorocitrate (FC). The responses were evaluated using immunocytochemistry, qPCR, and electrochemiluminescence. We found that SUP induced an inflammatory response characterized by increased interleukin (IL)-6 staining and increased expression of genes for IL-6, interferon (IFN)γ, and tumor necrosis factor (TNF)α along with genes associated with cell-to-cell communication such as C-C motif chemokine ligand-2 (CCL2). Inclusion of inhibitors attenuated some responses but failed to completely normalize all indications of response, highlighting the role of immunocompetent cellular crosstalk in regulating the inflammatory response. LTA and meso-DAP produced responses that shared characteristics with SUP but were not identical. Our results support a role for HF associated GP bacterial byproducts' ability to contribute to vagal inflammation and to engage signaling from nonneuronal cells.
Collapse
Affiliation(s)
- Carolina R Cawthon
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States
| | - Rebecca A Kirkland
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States
| | - Shreya Pandya
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States
| | - Nigel A Brinson
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States
| | - Claire B de La Serre
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States.
| |
Collapse
|
57
|
Treading water: mixed effects of high fat diet on mouse behavior in the forced swim test. Physiol Behav 2020; 223:112965. [DOI: 10.1016/j.physbeh.2020.112965] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 05/02/2020] [Accepted: 05/10/2020] [Indexed: 12/27/2022]
|
58
|
Espinoza L, Boychuk CR. Diabetes, and its treatment, as an effector of autonomic nervous system circuits and its functions. Curr Opin Pharmacol 2020; 54:18-26. [PMID: 32721846 DOI: 10.1016/j.coph.2020.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 06/18/2020] [Accepted: 06/22/2020] [Indexed: 12/24/2022]
Abstract
Diabetes increases the risk of cardiovascular complications, including heart failure, hypertension, and stroke. There is a strong involvement of autonomic dysfunction in individuals with diabetes that exhibit clinical manifestations of cardiovascular diseases (CVD). Still, the mechanisms by which diabetes and its treatments alter autonomic function and subsequently affect cardiovascular complications remain elusive. For this reason, understanding the brainstem circuits involved in sensing metabolic state(s) and enacting autonomic control of the cardiovascular system are important to develop more comprehensive therapies for individuals with diabetes at increased risk for CVD. We review how autonomic nervous system circuits change during these disease states and discuss their potential role in current pharmacotherapies that target diabetic states. Overall, this review proposes that the brainstem circuits provide an integrative sensorimotor network capable of responding to metabolic cues to regulate cardiovascular function and this network is modified by, and in turn affects, diabetes-induced CVD and its treatment.
Collapse
Affiliation(s)
- Liliana Espinoza
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, United States
| | - Carie R Boychuk
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, United States.
| |
Collapse
|
59
|
Kim JS, Kirkland RA, Lee SH, Cawthon CR, Rzepka KW, Minaya DM, de Lartigue G, Czaja K, de La Serre CB. Gut microbiota composition modulates inflammation and structure of the vagal afferent pathway. Physiol Behav 2020; 225:113082. [PMID: 32682966 DOI: 10.1016/j.physbeh.2020.113082] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 07/02/2020] [Accepted: 07/15/2020] [Indexed: 12/17/2022]
Abstract
Vagal afferent neurons (VAN), located in the nodose ganglion (NG) innervate the gut and terminate in the nucleus of solitary tract (NTS) in the brainstem. They are the primary sensory neurons integrating gut-derived signals to regulate meal size. Chronic high-fat diet (HFD) consumption impairs vagally mediated satiety, resulting in overfeeding. There is evidence that HFD consumption leads to alterations in both vagal nerve function and structural integrity. HFD also leads to marked gut microbiota dysbiosis; in rodent models, dysbiosis is sufficient to induce weight gain. In this study, we investigated the effect of microbiota dysbiosis on gut-brain vagal innervation independently of diet. To do so, we recolonized microbiota-depleted rats with gastrointestinal (GI) contents isolated from donor animals fed either a HFD (45 or 60% fat) or a low fat diet (LFD, 13% fat). We used two different depletion models while maintaining the animals on LFD: 1) conventionally raised Fischer and Wistar rats that underwent a depletion paradigm using an antibiotic cocktail and 2) germ free (GF) raised Fischer rats. Following recolonization, receiver animals were designated as ConvLF and ConvHF. Fecal samples were collected throughout these studies and analyzed via 16S Illumina sequencing. In both models, bacteria that were identified as characteristic of HFD were successfully transferred to recipient animals. Three weeks post-colonization, ConvHF rats showed significant increases in ionized calcium-binding adapter molecule-1 (Iba1) positive immune cells in the NG compared to ConvLF animals. Additionally, using isolectin B4 (IB4) staining to identify c-fibers, we found that, compared to ConvLF animals, ConvHF rats displayed decreased innervation at the level of the medial NTS; c-fibers at this level are believed to be primarily of vagal origin. This alteration in vagal structure was associated with a loss in satiety induced by the gut peptide cholecystokinin (CCK). Increased presence of immunocompetent Iba1+ cells along the gut-brain axis and alterations in NTS innervation were still evident in ConvHF rats compared to ConvLF animals 12 weeks post-colonization and were associated with increases in food intake and body weight (BW). We conclude from these data that microbiota dysbiosis can alter gut-brain vagal innervation, potentially via recruitment and/or activation of immune cells.
Collapse
Affiliation(s)
- J S Kim
- Dept. of Foods and Nutrition, USA
| | | | - S H Lee
- Dept. of Foods and Nutrition, USA
| | | | - K W Rzepka
- Dept. of Veterinary Biosciences and Diagnostic Imaging, University of Georgia, Athens, GA, USA
| | - D M Minaya
- Dept. of Veterinary Biosciences and Diagnostic Imaging, University of Georgia, Athens, GA, USA
| | - G de Lartigue
- Dept. of Pharmacodynamics, University of Florida, Gainesville, FL, USA
| | - K Czaja
- Dept. of Veterinary Biosciences and Diagnostic Imaging, University of Georgia, Athens, GA, USA
| | | |
Collapse
|
60
|
Cansell C, Stobbe K, Sanchez C, Le Thuc O, Mosser CA, Ben-Fradj S, Leredde J, Lebeaupin C, Debayle D, Fleuriot L, Brau F, Devaux N, Benani A, Audinat E, Blondeau N, Nahon JL, Rovère C. Dietary fat exacerbates postprandial hypothalamic inflammation involving glial fibrillary acidic protein-positive cells and microglia in male mice. Glia 2020; 69:42-60. [PMID: 32659044 DOI: 10.1002/glia.23882] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/15/2022]
Abstract
In humans, obesity is associated with brain inflammation, glial reactivity, and immune cells infiltration. Studies in rodents have shown that glial reactivity occurs within 24 hr of high-fat diet (HFD) consumption, long before obesity development, and takes place mainly in the hypothalamus (HT), a crucial brain structure for controlling body weight. Here, we sought to characterize the postprandial HT inflammatory response to 1, 3, and 6 hr of exposure to either a standard diet (SD) or HFD. HFD exposure increased gene expression of astrocyte and microglial markers (glial fibrillary acidic protein [GFAP] and Iba1, respectively) compared to SD-treated mice and induced morphological modifications of microglial cells in HT. This remodeling was associated with higher expression of inflammatory genes and differential regulation of hypothalamic neuropeptides involved in energy balance regulation. DREADD and PLX5622 technologies, used to modulate GFAP-positive or microglial cells activity, respectively, showed that both glial cell types are involved in hypothalamic postprandial inflammation, with their own specific kinetics and reactiveness to ingested foods. Thus, recurrent exacerbated postprandial inflammation in the brain might promote obesity and needs to be characterized to address this worldwide crisis.
Collapse
Affiliation(s)
- Céline Cansell
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Katharina Stobbe
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Clara Sanchez
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Ophélia Le Thuc
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Coralie-Anne Mosser
- Laboratory of Neurophysiology and New Microscopies, INSERM, Université Paris Descartes, Paris, France
| | - Selma Ben-Fradj
- CSGA, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| | - Joris Leredde
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | | | - Delphine Debayle
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Lucile Fleuriot
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Frédéric Brau
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Nadège Devaux
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Alexandre Benani
- CSGA, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| | - Etienne Audinat
- IGF, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Nicolas Blondeau
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Jean-Louis Nahon
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Carole Rovère
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| |
Collapse
|
61
|
Obesity Drives Delayed Infarct Expansion, Inflammation, and Distinct Gene Networks in a Mouse Stroke Model. Transl Stroke Res 2020; 12:331-346. [PMID: 32588199 DOI: 10.1007/s12975-020-00826-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/11/2020] [Accepted: 06/03/2020] [Indexed: 01/11/2023]
Abstract
Obesity is associated with chronic peripheral inflammation, is a risk factor for stroke, and causes increased infarct sizes. To characterize how obesity increases infarct size, we fed a high-fat diet to wild-type C57BL/6J mice for either 6 weeks or 15 weeks and then induced distal middle cerebral artery strokes. We found that infarct expansion happened late after stroke. There were no differences in cortical neuroinflammation (astrogliosis, microgliosis, or pro-inflammatory cytokines) either prior to or 10 h after stroke, and also no differences in stroke size at 10 h. However, by 3 days after stroke, animals fed a high-fat diet had a dramatic increase in microgliosis and astrogliosis that was associated with larger strokes and worsened functional recovery. RNA sequencing revealed a dramatic increase in inflammatory genes in the high-fat diet-fed animals 3 days after stroke that were not present prior to stroke. Genetic pathways unique to diet-induced obesity were primarily related to adaptive immunity, extracellular matrix components, cell migration, and vasculogenesis. The late appearance of neuroinflammation and infarct expansion indicates that there may be a therapeutic window between 10 and 36 h after stroke where inflammation and obesity-specific transcriptional programs could be targeted to improve outcomes in people with obesity and stroke.
Collapse
|
62
|
Potential therapeutic applications of the gut microbiome in obesity: from brain function to body detoxification. Int J Obes (Lond) 2020; 44:1818-1831. [PMID: 32523034 DOI: 10.1038/s41366-020-0618-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 05/04/2020] [Accepted: 05/22/2020] [Indexed: 02/08/2023]
Abstract
The prevalence of obesity is rising every year and associated comorbidities such as cardiovascular diseases are among the leading causes of death worldwide. The gut microbiota has recently emerged as a potential target for therapeutic applications to prevent and treat those comorbidities. In this review, we focus on three conditions related to obesity in which the use of gut microbiota modulators could have benefits; mood disorders, eating behaviors, and body detoxification of persistent organic pollutants (POPs). On one hand, modulation of gut-derived signals to the brain in a context of obesity is involved in the development of neuroinflammation and can subsequently alter behaviors. An altered gut microbiome could change these signals and alleviate their consequences. On the other hand, obesity is associated with an increased accumulation of lipophilic contaminants, such as POPs. Targeting the microbiota could help body detoxication by reducing bioavailability, enhancing degradation by bioremediation or their excretion through the enterohepatic circulation. Thus, a supplementation of prebiotics, probiotics, or synbiotics could represent a complementary strategy to current ones, such as medication and lifestyle modifications, to decrease depression, alter eating behaviors, and lower body burden of pollutants considering the actual obesity epidemic our society is facing.
Collapse
|
63
|
Consumption of a high energy density diet triggers microbiota dysbiosis, hepatic lipidosis, and microglia activation in the nucleus of the solitary tract in rats. Nutr Diabetes 2020; 10:20. [PMID: 32518225 PMCID: PMC7283362 DOI: 10.1038/s41387-020-0119-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/30/2020] [Accepted: 05/06/2020] [Indexed: 02/06/2023] Open
Abstract
Introduction Obesity is a multifactorial chronic inflammatory disease. Consumption of high energy density (HED) diets is associated with hyperphagia, increased body weight and body fat accumulation, and obesity. Our lab has previously shown that short-term (4 weeks) consumption of a HED diet triggers gut microbiota dysbiosis, gut inflammation, and reorganization of the gut-brain vagal communication. Objetives The aim of this study was to investigate the effect of long-term (6 months) consumption of HED diet on body composition, gut microbiome, hepatocellular lipidosis, microglia activation in the nucleus of the solitary tract, and systemic inflammation. Methods Male Sprague–Dawley rats were fed a low energy density (LED) diet for 2 weeks and then switched to a HED diet for 26 weeks. Twenty-four-hour food intake, body weight, and body composition were measured twice a week. Blood serum and fecal samples were collected at baseline, 1, 4, 8, and 26 weeks after introduction of the HED diet. Serum samples were used to measure insulin, leptin, and inflammatory cytokines using Enzyme-linked Immunosorbent Assay. Fecal samples were assessed for 16 S rRNA genome sequencing. Results HED diet induced microbiota dysbiosis within a week of introducing the diet. In addition, there was significant microglia activation in the intermediate NTS and marked hepatic lipidosis after 4 weeks of HED diet. We further observed changes in the serum cytokine profile after 26 weeks of HED feeding. Conclusions These data suggest that microbiota dysbiosis is the first response of the organism to HED diets, followed by increased liver fat accumulation, microglia activation in the brain, and circulating levels of inflammatory markers. To our knowledge, this is the first study to present longitudinal and cross-sectional results on effect of long-term consumption of HED diets on all these parameters in a single cohort of animals.
Collapse
|
64
|
Lee TJ, Hargrave SL, Kinzig KP. Dual functions of CNS inflammation in food intake and metabolic regulation. Brain Res 2020; 1740:146859. [PMID: 32353432 DOI: 10.1016/j.brainres.2020.146859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 04/08/2020] [Accepted: 04/25/2020] [Indexed: 01/22/2023]
Abstract
Western diet (WD) consumption induces chronic mild inflammation in the hypothalamus. However, metabolic consequences of increased hypothalamic inflammatory cytokines remain unclear. This research first aimed to examine whether increased proinflammatory cytokines in the brain influenced feeding or metabolism. Rats that received an intracerebroventricular third ventricle injection (i3vt) of 0.5 pg TNFα daily for six days consumed significantly more calories than saline-injected rats, with no differences between treatment groups in terms of body weight, blood triglycerides nor glucose regulation. Continuously infusing TNFα for three weeks decreased hepatic fatty acid synthase (FAS) and increased body weight and the epididymal adipose sterol regulatory element-binding protein 1c (SREBP-1c) gene expression. Differences were not due to food intake nor voluntary wheel running activity. The second aim of this research was to examine whether inhibition of inflammation signaling in the brain at early stage of switching from chow to WD would affect diet-induced obesity development. WD-fed rats with i3vt NFκB inhibitor had greater caloric intake than rats given i3vt saline. These studies suggest elevated inflammatory cytokines in the brain induce food intake acutely and favor fat storage and weight gain in the long term. However, in the early stage of WD consumption, hypothalamic inflammatory signaling inhibits caloric intake and may serve as a warning signal of energy imbalance.
Collapse
Affiliation(s)
- Tien-Jui Lee
- Department of Psychological Sciences, Purdue University, 703 Third Street, West Lafayette, IN 47907, United States.
| | - Sara L Hargrave
- Department of Psychological Sciences, Purdue University, 703 Third Street, West Lafayette, IN 47907, United States
| | - Kimberly P Kinzig
- Department of Psychological Sciences, Purdue University, 703 Third Street, West Lafayette, IN 47907, United States.
| |
Collapse
|
65
|
Poon K. Behavioral Feeding Circuit: Dietary Fat-Induced Effects of Inflammatory Mediators in the Hypothalamus. Front Endocrinol (Lausanne) 2020; 11:591559. [PMID: 33324346 PMCID: PMC7726204 DOI: 10.3389/fendo.2020.591559] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/23/2020] [Indexed: 12/19/2022] Open
Abstract
Excessive dietary fat intake has extensive impacts on several physiological systems and can lead to metabolic and nonmetabolic disease. In animal models of ingestion, exposure to a high fat diet during pregnancy predisposes offspring to increase intake of dietary fat and causes increase in weight gain that can lead to obesity, and without intervention, these physiological and behavioral consequences can persist for several generations. The hypothalamus is a region of the brain that responds to physiological hunger and fullness and contains orexigenic neuropeptide systems that have long been associated with dietary fat intake. The past fifteen years of research show that prenatal exposure to a high fat diet increases neurogenesis of these neuropeptide systems in offspring brain and are correlated to behavioral changes that induce a pro-consummatory and obesogenic phenotype. Current research has uncovered several potential molecular mechanisms by which excessive dietary fat alters the hypothalamus and involve dietary fatty acids, the immune system, gut microbiota, and transcriptional and epigenetic changes. This review will examine the current knowledge of dietary fat-associated changes in the hypothalamus and the potential pathways involved in modifying the development of orexigenic peptide neurons that lead to changes in ingestive behavior, with a special emphasis on inflammation by chemokines.
Collapse
|
66
|
Reduced central and peripheral inflammatory responses and increased mitochondrial activity contribute to diet-induced obesity resistance in WSB/EiJ mice. Sci Rep 2019; 9:19696. [PMID: 31873127 PMCID: PMC6928236 DOI: 10.1038/s41598-019-56051-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 11/27/2019] [Indexed: 12/13/2022] Open
Abstract
Energy imbalance due to excess of calories is considered to be a major player in the current worldwide obesity pandemic and could be accompanied by systemic and central inflammation and mitochondrial dysfunctions. This hypothesis was tested by comparing the wild-derived diet-induced obesity- (DIO-) resistant mouse strain WSB/EiJ to the obesity-prone C57BL/6J strain. We analysed circulating and hypothalamic markers of inflammatory status and hypothalamic mitochondrial activity in both strains exposed to high-fat diet (HFD). We further analysed the regulations of hypothalamic genes involved in inflammation and mitochondrial pathways by high throughput microfluidic qPCR on RNA extracted from laser micro-dissected arcuate (ARC) and paraventricular (PVN) hypothalamic nuclei. HFD induced increased body weight gain, circulating levels of leptin, cholesterol, HDL and LDL in C57BL/6J whereas WSB/EiJ mice displayed a lower inflammatory status, both peripherally (lower levels of circulating cytokines) and centrally (less activated microglia in the hypothalamus) as well as more reactive mitochondria in the hypothalamus. The gene expression data analysis allowed identifying strain-specific hypothalamic metabolic pathways involved in the respective responses to HFD. Our results point to the involvement of hypothalamic inflammatory and mitochondrial pathways as key factors in the control of energy homeostasis and the resistance to DIO.
Collapse
|
67
|
Ntambi JM. Highlighting inflammation and lipid metabolism. Biochem Biophys Res Commun 2019; 520:688-689. [DOI: 10.1016/j.bbrc.2019.10.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 10/01/2019] [Indexed: 11/29/2022]
|
68
|
Klinedinst BS, Pappas C, Le S, Yu S, Wang Q, Wang L, Allenspach-Jorn K, Mochel JP, Willette AA. Aging-related changes in fluid intelligence, muscle and adipose mass, and sex-specific immunologic mediation: A longitudinal UK Biobank study. Brain Behav Immun 2019; 82:396-405. [PMID: 31513875 PMCID: PMC7755032 DOI: 10.1016/j.bbi.2019.09.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/07/2019] [Accepted: 09/08/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Obesity in midlife and early late-life is associated with worse normal cognitive aging. Dual-energy X-ray absorptiometry (DEXA) suggests that visceral adipose mass (VAM) plays a predominant role, whereas non-visceral adipose mass (NVAM) and lean muscle mass (LMM) have shown conflicting relationships. It is unknown how longitudinal, cognitive changes in age-sensitive domains like fluid intelligence (FI) correspond to VAM, NVAM, and LMM in women and men. Furthermore, changes over time in blood leukocyte sub-populations may partially or fully account for sex-specific associations. METHODS Data on 4431 late middle-aged, cognitively unimpaired adults (mean = 64.5 y) was obtained from the UK Biobank prospective cohort across 22 centers. FI scores, blood leukocyte counts, and covariates (age, social class, education) were measured at three 2-year intervals over 6 years. DEXA collection overlapped with these intervals. Sex-stratified growth curves, structural equations, and Preacher-Hayes mediation were used to estimate direct and indirect effects. β-weights were standardized. RESULTS More LMM predicted gains in FI scores among women (β = 0.130, p < .001) and men (β = 0.089, p < .001). Conversely, more VAM and NVAM independently predicted FI decline equally among sexes (e.g., NVAM: women: β = -0.082, p < .001; men: β = -0.076, p < .001). Among women, FI associations were fully mediated by higher eosinophil counts via VAM (λ = 30.8%, p = .028) and lower lymphocyte counts via LMM (λ = 69.2%, p = .021). Among men, FI associations were partially mediated by lower basophils counts via LMM (λ = 4.5%, p = .042) and higher counts via VAM (λ = 50%, p = .037). CONCLUSION The proportion of LMM and VAM equally influenced male FI changes over 6 years, whereas higher LMM among women appeared to more strongly influence. FI changes. Leukocyte counts strongly mediated VAM- and LMM-related FI changes in a sex-specific manner, but not for NVAM. For clinical translation, exercise studies in older adults may benefit from assessing sex-specific values of DEXA-based tissue mass, FI, and leukocyte sub-populations to gauge potential cognitive benefits of less VAM and more LMM.
Collapse
Affiliation(s)
- Brandon S. Klinedinst
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA,Neuroscience Graduate Program, Iowa State University, Ames, IA, USA
| | - Colleen Pappas
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA
| | - Scott Le
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA,Interdisciplinary Graduate Studies Program, Iowa State University, Ames, IA, USA
| | - Shan Yu
- Department of Statistics, Iowa State University, Ames, IA, USA
| | - Qian Wang
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA,Neuroscience Graduate Program, Iowa State University, Ames, IA, USA
| | - Li Wang
- Department of Statistics, Iowa State University, Ames, IA, USA
| | | | | | - Auriel A. Willette
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA,Neuroscience Graduate Program, Iowa State University, Ames, IA, USA,Interdisciplinary Graduate Studies Program, Iowa State University, Ames, IA, USA,Department of Biomedical Sciences, Iowa State University, Ames, IA, USA,Department of Neurology, University of Iowa, Iowa City, USA,Send Correspondence to: Auriel A. Willette, 1109 HNSB, 2302 Osborn Drive, Ames, IA 50011-1078, Phone: (515) 294-3110,
| |
Collapse
|
69
|
Peixoto TC, Moura EG, Oliveira E, Younes-Rapozo V, Soares PN, Rodrigues VST, Torsoni MA, Torsoni AS, Manhães AC, Lisboa PC. Hypothalamic Neuropeptides Expression and Hypothalamic Inflammation in Adult Rats that Were Exposed to Tobacco Smoke during Breastfeeding: Sex-Related Differences. Neuroscience 2019; 418:69-81. [PMID: 31487543 DOI: 10.1016/j.neuroscience.2019.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 02/06/2023]
Abstract
The hypothalamus controls food intake and energy expenditure. In rats, maternal exposure to nicotine during breastfeeding alters the hypothalamic circuitry of the adult offspring, resulting in leptin resistance, neuropeptides changes and gliosis. Tobacco smoke exposure during lactation causes greater adiposity, hyperphagia and hyperleptinemia in the adult progeny. To understand the central mechanisms underlying the obese phenotype of adult rats that were directly and indirectly exposed to cigarette smoke during lactation, we investigated leptin signaling, orexigenic and anorexigenic neuropeptides expression, as well as astrocyte and microglia markers in hypothalamus. From postnatal day (PND) 3 to 21, Wistar lactating rat dams and their pups were divided into two groups: SE, smoke-exposed in a cigarette-smoking machine (four times/day); Crtl, exposed to filtered air. Offspring of both sexes were euthanized at PND180. The leptin pathway was not altered in SE animals from both sexes. SE males showed increased NPY (arcuate nucleus, ARC), CRH (paraventricular nucleus, PVN), as well as higher GFAP fiber density (ARC and PVN) and IL6 protein content. TRH (PVN) immunohistochemistry was reduced. SE females had lower CART-positive cells (ARC) and lower α-MSH immunostaining intensity (PVN and lateral hypothalamus), with no change of GFAP or IL-6. The protein contents of CX3CR1 (marker of activated microglia) and α7nAChR (anti-inflammatory marker) were not altered in both SE males and females. Neonatal cigarette smoke is deleterious to the hypothalamic circuitry, inducing changes in energy homeostasis favoring hyperphagia and decreased energy expenditure at adulthood in both sexes; however sex-dependent mechanisms were observed.
Collapse
Affiliation(s)
- T C Peixoto
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - E G Moura
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - E Oliveira
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - V Younes-Rapozo
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - P N Soares
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - V S T Rodrigues
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - M A Torsoni
- Faculdade de Ciências Aplicadas, Universidade Estadual de Campinas, Limeira, Brazil
| | - A S Torsoni
- Faculdade de Ciências Aplicadas, Universidade Estadual de Campinas, Limeira, Brazil
| | - A C Manhães
- Laboratório de Neurofisiologia, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - P C Lisboa
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
70
|
Rey M, Kruse MS, Magrini-Huamán RN, Coirini H. High-Fat Diets and LXRs Expression in Rat Liver and Hypothalamus. Cell Mol Neurobiol 2019; 39:963-974. [PMID: 31161476 PMCID: PMC11457829 DOI: 10.1007/s10571-019-00692-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/25/2019] [Indexed: 12/25/2022]
Abstract
Disturbances on lipid metabolism are associated with health disorders. High-fat diets (HFDs) consumption promotes cardiovascular and neurodegenerative diseases where cholesterol plays an important role. Among regulators of this steroid homeostasis, the liver X receptors (LXRs) induce genes that protect cells from cholesterol overload. We previously described how both hypothalamic LXRα and LXRβ are sensitive to a high-fructose diet, suggesting that these receptors trigger responses related to control of energy and food intake. The present work's main objective was to study the effect of different HFDs on LXRs expression (in hypothalamus and liver), and lipid profile. Male rats received control diet (CD), HFD1 (CD + bovine fat (BF)), HFD2 (CD + BF + cholic acid (CA)), HFD3 (CD + BF + cholesterol), or HFD4 (CD + BF + CA + cholesterol) for different time periods. Hypothalamic LXRβ, both hepatic LXRs subtypes, and total cholesterol (TC) raised after 2 weeks of HFDs. Four and 8 weeks of HFD3 and HFD4 increased the LXRs subtypes in both tissues and TC levels. Only HFD4 reduced triglycerides (TG) levels after 2 and 8 weeks. The TC and TG values correlated significantly with LXRs expression only in rats fed with HFD4. These data add relevant information about how diet composition can produce different scales of hypercholesterolemia states accompanied with central and peripheral changes in the LXRs expression.
Collapse
Affiliation(s)
- Mariana Rey
- Laboratorio de Neurobiologia, Instituto de Biologia y Medicina Experimental (IBYME-CONICET), Vuelta de Obligado 2490, C1428ADN, Ciudad Autonoma de Buenos Aires, Buenos Aires, Argentina
| | - María Sol Kruse
- Laboratorio de Neurobiologia, Instituto de Biologia y Medicina Experimental (IBYME-CONICET), Vuelta de Obligado 2490, C1428ADN, Ciudad Autonoma de Buenos Aires, Buenos Aires, Argentina
| | - Rocío Nahimé Magrini-Huamán
- Laboratorio de Neurobiologia, Instituto de Biologia y Medicina Experimental (IBYME-CONICET), Vuelta de Obligado 2490, C1428ADN, Ciudad Autonoma de Buenos Aires, Buenos Aires, Argentina
- Facultad de Ingenieria, Instituto de Biotecnologia, Universidad Nacional de San Juan, Av. Libertador Gral. San Martín 1109, J5400ARL, San Juan, Argentina
- Facultad de Ciencias Medicas, Universidad Catolica de Cuyo, Av. José Ignacio de la Roza 1516, Rivadavia, J5400, San Juan, Argentina
| | - Héctor Coirini
- Laboratorio de Neurobiologia, Instituto de Biologia y Medicina Experimental (IBYME-CONICET), Vuelta de Obligado 2490, C1428ADN, Ciudad Autonoma de Buenos Aires, Buenos Aires, Argentina.
- Facultad de Ciencias Medicas, Universidad Catolica de Cuyo, Av. José Ignacio de la Roza 1516, Rivadavia, J5400, San Juan, Argentina.
- Departamento de Bioquimica Humana, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, 5to Piso, C1121ABG, Ciudad Autonoma de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
71
|
Noronha SSR, Lima PM, Campos GSV, Chírico MTT, Abreu AR, Figueiredo AB, Silva FCS, Chianca DA, Lowry CA, De Menezes RCA. Association of high-fat diet with neuroinflammation, anxiety-like defensive behavioral responses, and altered thermoregulatory responses in male rats. Brain Behav Immun 2019; 80:500-511. [PMID: 31022457 DOI: 10.1016/j.bbi.2019.04.030] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/18/2019] [Accepted: 04/22/2019] [Indexed: 12/21/2022] Open
Abstract
Overweight and obesity are a worldwide pandemic affecting billions of people. These conditions have been associated with a chronic low-grade inflammatory state that is recognized as a risk factor for a range of somatic diseases as well as neurodevelopmental disorders, anxiety disorders, trauma- and stressor-related disorders, and affective disorders. We previously reported that the ingestion of a high-fat diet (HFD; 45% fat kcal/g) for nine weeks was capable of inducing obesity in rats in association with increased reactivity to stress and increased anxiety-related defensive behavior. In this study, we conducted a nine-week diet protocol to induce obesity in rats, followed by investigation of anxiety-related defensive behavioral responses using the elevated T-maze (ETM), numbers of FOS-immunoreactive cells after exposure of rats to the avoidance or escape task of the ETM, and neuroinflammatory cytokine expression in hypothalamic and amygdaloid nuclei. In addition, we investigated stress-induced cutaneous thermoregulatory responses during exposure to an open-field (OF). Here we demonstrated that nine weeks of HFD intake induced obesity, in association with increased abdominal fat pad weight, increased anxiety-related defensive behavioral responses, and increased proinflammatory cytokines in hypothalamic and amygdaloid nuclei. In addition, HFD exposure altered avoidance- or escape task-induced FOS-immunoreactivity within brain structures involved in control of neuroendocrine, autonomic, and behavioral responses to aversive stimuli, including the basolateral amygdala (BLA) and dorsomedial (DMH), paraventricular (PVN) and ventromedial (VMH) hypothalamic nuclei. Furthermore, rats exposed to HFD, relative to control diet-fed rats, responded with increased tail skin temperature at baseline and throughout exposure to an open-field apparatus. These data are consistent with the hypothesis that HFD induces neuroinflammation, alters excitability of brain nuclei controlling neuroendocrine, autonomic, and behavioral responses to stressful stimuli, and enhances stress reactivity and anxiety-like defensive behavioral responses.
Collapse
Affiliation(s)
- S S R Noronha
- Department of Biological Science, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil; Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - P M Lima
- Department of Biological Science, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil
| | - G S V Campos
- Department of Biological Science, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil
| | - M T T Chírico
- Department of Biological Science, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil
| | - A R Abreu
- Department of Biological Science, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil
| | - A B Figueiredo
- Department of Biological Science, Laboratory of Immunoparasitology, University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil
| | - F C S Silva
- Department of Biological Science, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil
| | - D A Chianca
- Department of Biological Science, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil
| | - C A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; Center for Neuroscience and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO 80309, USA; Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO 80045, USA; Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Aurora, CO 80045, USA
| | - R C A De Menezes
- Department of Biological Science, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG 35400-000, Brazil.
| |
Collapse
|
72
|
Interscapular and Perivascular Brown Adipose Tissue Respond Differently to a Short-Term High-Fat Diet. Nutrients 2019; 11:nu11051065. [PMID: 31086124 PMCID: PMC6566556 DOI: 10.3390/nu11051065] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 12/18/2022] Open
Abstract
Brown adipose tissue (BAT) function may depend on its anatomical location and developmental origin. Interscapular BAT (iBAT) regulates acute macronutrient metabolism, whilst perivascular BAT (PVAT) regulates vascular function. Although phenotypically similar, whether these depots respond differently to acute nutrient excess is unclear. Given their distinct anatomical locations and developmental origins and we hypothesised that iBAT and PVAT would respond differently to brief period of nutrient excess. Sprague-Dawley rats aged 12 weeks (n=12) were fed either a standard (10% fat, n=6) or high fat diet (HFD: 45% fat, n=6) for 72h and housed at thermoneutrality. Following an assessment of whole body physiology, fat was collected from both depots for analysis of gene expression and the proteome. HFD consumption for 72h induced rapid weight gain (c. 2.6%) and reduced serum non-esterified fatty acids (NEFA) with no change in either total adipose or depot mass. In iBAT, an upregulation of genes involved in insulin signalling and lipid metabolism was accompanied by enrichment of lipid-related processes and functions, plus glucagon and peroxisome proliferator-activated receptor (PPAR) signalling pathways. In PVAT, HFD induced a pronounced down-regulation of multiple metabolic pathways which was accompanied with increased abundance of proteins involved in apoptosis (e.g., Hdgf and Ywaq) and toll-like receptor signalling (Ube2n). There was also an enrichment of DNA-related processes and functions (e.g., nucleosome assembly and histone exchange) and RNA degradation and cell adhesion pathways. In conclusion, we show that iBAT and PVAT elicit divergent responses to short-term nutrient excess highlighting early adaptations in these depots before changes in fat mass.
Collapse
|
73
|
Souza ACP, Souza CM, Amaral CL, Lemes SF, Santucci LF, Milanski M, Torsoni AS, Torsoni MA. Short-Term High-Fat Diet Consumption Reduces Hypothalamic Expression of the Nicotinic Acetylcholine Receptor α7 Subunit (α7nAChR) and Affects the Anti-inflammatory Response in a Mouse Model of Sepsis. Front Immunol 2019; 10:565. [PMID: 30967878 PMCID: PMC6438922 DOI: 10.3389/fimmu.2019.00565] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 03/04/2019] [Indexed: 01/01/2023] Open
Abstract
Sepsis is one of the leading causes of death in hospitalized patients and the chronic and low-grade inflammation observed in obesity seems to worsen susceptibility and morbidity of infections. However, little is known with respect to a short-term high-fat diet (HFD) and its role in the development of sepsis. Here, we show for the first time, that short-term HFD consumption impairs early nicotinic acetylcholine receptor α7 subunit (α7nAChR)- mediated signaling, one of the major components of the cholinergic anti-inflammatory pathway, with a focus on hypothalamic inflammation and innate immune response. Mice were randomized to a HFD or standard chow (SC) for 3 days, and sepsis was subsequently induced by a lethal intraperitoneal (i.p.) injection of lipopolysaccharide (LPS) or by cecal ligation and puncture (CLP) surgery. In a separate experiment, both groups received LPS (i.p.) or LPS (i.p.) in conjunction with the selective α7nAChR agonist, PNU-282987 (i.p. or intracerebroventricular; i.c.v.), and were sacrificed 2 h after the challenge. Short-term HFD consumption significantly reduced the α7nAChR mRNA and protein levels in the hypothalamus and liver (p < 0.05). Immunofluorescence microscopy demonstrated lower cholinergic receptor nicotinic α7 subunit (α7nAChR)+ cells in the arcuate nucleus (ARC) (α7nAChR+ cells in SC = 216 and HFD = 84) and increased F4/80+ cells in the ARC (2.6-fold) and median eminence (ME) (1.6-fold), which can contribute to neuronal damage. Glial fibrillary acidic protein (GFAP)+ cells and neuronal nuclear antigen (NeuN)+ cells were also increased following consumption of HFD. The HFD-fed mice died quickly after a lethal dose of LPS or following CLP surgery (2-fold compared with SC). The LPS challenge raised most cytokine levels in both groups; however, higher levels of TNF-α (Spleen and liver), IL-1β and IL-6 (in all tissues evaluated) were observed in HFD-fed mice. Moreover, PNU-282987 administration (i.p. or i.c.v.) reduced the levels of inflammatory markers in the hypothalamus following LPS injection. Nevertheless, when the i.c.v. injection of PNU-282987 was performed the anti-inflammatory effect was much smaller in HFD-fed mice than SC-fed mice. Here, we provide evidence that a short-term HFD impairs early α7nAChR expression in central and peripheral tissues, contributing to a higher probability of death in sepsis.
Collapse
Affiliation(s)
- Anelise Cristina Parras Souza
- School of Applied Sciences, University of Campinas, Limeira, Brazil.,Obesity and Comorbidities Research Center, State University of Campinas, Limeira, Brazil
| | - Camilla Mendes Souza
- School of Applied Sciences, University of Campinas, Limeira, Brazil.,Obesity and Comorbidities Research Center, State University of Campinas, Limeira, Brazil
| | - Camila Libardi Amaral
- School of Applied Sciences, University of Campinas, Limeira, Brazil.,Obesity and Comorbidities Research Center, State University of Campinas, Limeira, Brazil
| | - Simone Ferreira Lemes
- School of Applied Sciences, University of Campinas, Limeira, Brazil.,Obesity and Comorbidities Research Center, State University of Campinas, Limeira, Brazil
| | - Leticia Foglia Santucci
- School of Applied Sciences, University of Campinas, Limeira, Brazil.,Obesity and Comorbidities Research Center, State University of Campinas, Limeira, Brazil
| | - Marciane Milanski
- School of Applied Sciences, University of Campinas, Limeira, Brazil.,Obesity and Comorbidities Research Center, State University of Campinas, Limeira, Brazil
| | - Adriana Souza Torsoni
- School of Applied Sciences, University of Campinas, Limeira, Brazil.,Obesity and Comorbidities Research Center, State University of Campinas, Limeira, Brazil
| | - Marcio Alberto Torsoni
- School of Applied Sciences, University of Campinas, Limeira, Brazil.,Obesity and Comorbidities Research Center, State University of Campinas, Limeira, Brazil
| |
Collapse
|
74
|
Okada T, Mita Y, Sakoda H, Nakazato M. Impaired adaptation of energy intake induces severe obesity in aged mice on a high-fat diet. Physiol Rep 2019; 7:e13989. [PMID: 30706678 PMCID: PMC6356180 DOI: 10.14814/phy2.13989] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/27/2018] [Accepted: 12/29/2018] [Indexed: 01/13/2023] Open
Abstract
High-fat diet (HFD) feeding induces inflammation in various tissues, including the nodose ganglion and hypothalamus, resulting in obesity and metabolic disorders. In this study, we investigated the effect of short-term HFD on aged and young mice. Aged mice easily gained weight during short-term HFD feeding, and required many days to adapt their energy intake. One-day HFD in aged mice induced inflammation in the distal colon, but not in the nodose ganglion or hypothalamus. The anorexic effect of glucagon-like peptide-1 (GLP-1) was attenuated in aged mice. Intraperitoneal administration of GLP-1 did not induce expression of genes that regulate feeding in the hypothalamus of aged mice. mRNA expression of the gene encoding the GLP-1 receptor (Glp1r) in the nodose ganglion was significantly lower in aged mice than in young mice. Our findings suggest that adaptation of energy intake regulation was attenuated in aged mice, causing them to become obese in response to short-term HFD feeding.
Collapse
Affiliation(s)
- Tadashi Okada
- Division of Neurology, Respirology, Endocrinology and MetabolismDepartment of Internal MedicineFaculty of MedicineUniversity of MiyazakiMiyazakiJapan
| | - Yuichiro Mita
- Division of Neurology, Respirology, Endocrinology and MetabolismDepartment of Internal MedicineFaculty of MedicineUniversity of MiyazakiMiyazakiJapan
| | - Hideyuki Sakoda
- Division of Neurology, Respirology, Endocrinology and MetabolismDepartment of Internal MedicineFaculty of MedicineUniversity of MiyazakiMiyazakiJapan
| | - Masamitsu Nakazato
- Division of Neurology, Respirology, Endocrinology and MetabolismDepartment of Internal MedicineFaculty of MedicineUniversity of MiyazakiMiyazakiJapan
- CREST (Japan) Agency for Medical Research and Development (A‐MED)Chiyoda‐kuTokyoJapan
| |
Collapse
|
75
|
Clyburn C, Browning KN. Role of astroglia in diet-induced central neuroplasticity. J Neurophysiol 2019; 121:1195-1206. [PMID: 30699056 DOI: 10.1152/jn.00823.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Obesity, characterized by increased adiposity that develops when energy intake outweighs expenditure, is rapidly becoming a serious health crisis that affects millions of people worldwide and is associated with severe comorbid disorders including hypertension, cardiovascular disease, and type II diabetes. Obesity is also associated with the dysregulation of central neurocircuits involved in the control of autonomic, metabolic, and cognitive functions. Systemic inflammation associated with diet-induced obesity (DIO) has been proposed to be responsible for the development of these comorbidities as well as the dysregulation of central neurocircuits. A growing body of evidence suggests, however, that exposure to a high-fat diet (HFD) may cause neuroinflammation and astroglial activation even before systemic inflammation develops, which may be sufficient to cause dysregulation of central neurocircuits involved in energy homeostasis before the development of obesity. The purpose of this review is to summarize the current literature exploring astroglial-dependent modulation of central circuits following exposure to HFD and DIO, including not only dysregulation of neurocircuits involved in energy homeostasis and feeding behavior, but also the dysregulation of learning, memory, mood, and reward pathways.
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
|
76
|
Lizarbe B, Soares AF, Larsson S, Duarte JMN. Neurochemical Modifications in the Hippocampus, Cortex and Hypothalamus of Mice Exposed to Long-Term High-Fat Diet. Front Neurosci 2019; 12:985. [PMID: 30670942 PMCID: PMC6331468 DOI: 10.3389/fnins.2018.00985] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 12/10/2018] [Indexed: 11/13/2022] Open
Abstract
Metabolic syndrome and diabetes impact brain function and metabolism. While it is well established that rodents exposed to diets rich in saturated fat develop brain dysfunction, contrasting results abound in the literature, likely as result of exposure to different high-fat diet (HFD) compositions and for varied periods of time. In the present study, we investigated alterations of hippocampal-dependent spatial memory by measuring Y-maze spontaneous alternation, metabolic profiles of the hippocampus, cortex and hypothalamus by 1H magnetic resonance spectroscopy (MRS), and levels of proteins specific to synaptic and glial compartments in mice exposed for 6 months to different amounts of fat (10, 45, or 60% of total energy intake). Increasing the dietary amount of fat from 10 to 45% or 60% resulted in obesity accompanied by increased leptin, fasting blood glucose and insulin, and reduced glucose tolerance. In comparison to controls (10%-fat), only mice fed the 60%-fat diet showed increased fed glycemia, as well as plasma corticosterone that has a major impact on brain function. HFD-induced metabolic profile modifications measured by 1H MRS were observed across the three brain areas in mice exposed to 60%- but not 45%-fat diet, while both HFD groups displayed impaired hippocampal-dependent memory. HFD also affected systems involved in neuro- or gliotransmission in the hippocampus. Namely, relative to controls, 60%-fat-fed mice showed reduced SNAP-25, PSD-95 and syntaxin-4 immunoreactivity, while 45%-fat-fed mice showed reduced gephyrin and syntaxin-4 immunoreactivity. For both HFD levels, reductions of the vesicular glutamate transporter vGlut1 and levels of the vesicular GABA transporter were observed in the hippocampus and hypothalamus, relative to controls. Immunoreactivity against GFAP and/or Iba-1 in the hypothalamus was higher in mice exposed to HFD than controls, suggesting occurrence of gliosis. We conclude that different levels of dietary fat result in distinct neurochemical alterations in the brain.
Collapse
Affiliation(s)
- Blanca Lizarbe
- Laboratory for Functional and Metabolic Imaging, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid, Madrid, Spain
| | - Ana Francisca Soares
- Laboratory for Functional and Metabolic Imaging, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Sara Larsson
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - João M N Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
77
|
Milanova IV, Kalsbeek MJT, Wang XL, Korpel NL, Stenvers DJ, Wolff SEC, de Goede P, Heijboer AC, Fliers E, la Fleur SE, Kalsbeek A, Yi CX. Diet-Induced Obesity Disturbs Microglial Immunometabolism in a Time-of-Day Manner. Front Endocrinol (Lausanne) 2019; 10:424. [PMID: 31316470 PMCID: PMC6611391 DOI: 10.3389/fendo.2019.00424] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 06/12/2019] [Indexed: 12/16/2022] Open
Abstract
Background: Disturbance of immunometabolic signaling is a key process involved in the progression of obesity. Microglia-the resident immune cells in the brain, initiate local immune responses. It is known that hypercaloric diets lead to microglial activation. Previously, we observed that hypothalamic microglial cells from mice fed high-fat diet (HFD) lose their day/night rhythm and are constantly activated. However, little is known about daily rhythmicity in microglial circadian, immune and metabolic functions, either in lean or obese conditions. Therefore, we hypothesized that HFD disturbs microglial immunometabolism in a day/night-dependent manner. Methods: Obesity was induced in Wistar rats by feeding them HFD ad libitum for the duration of 8 weeks. Microglia were isolated from HFD- and chow-fed control animals at six time points during 24 h [every 4 h starting 2 h after lights on, i.e., Zeitgeber Time 2 (ZT2)]. Gene expression was evaluated using quantitative RT-PCR. JTK_Cycle software was used to estimate daily rhythmicity. Statistical analysis was performed with two-way ANOVA test. Results: Consumption of the obesogenic diet resulted in a 40 g significantly higher body weight gain in week 8, compared to chow diet (p < 0.0001), associated with increased adiposity. We observed significant rhythmicity of circadian clock genes in microglia under chow conditions, which was partially lost in diet-induced obesity (DIO). Microglial immune gene expression also showed time-of-day differences, which were disrupted in HFD-fed animals. Microglia responded to the obesogenic conditions by a shift of substrate utilization with decreased glutamate and glucose metabolism in the active period of the animals, and an overall increase of lipid metabolism, as indicated by gene expression evaluation. Additionally, data on mitochondria bioenergetics and dynamics suggested an increased energy production in microglia during the inactive period on HFD. Finally, evaluation of monocyte functional gene expression showed small or absent effect of HFD on peripheral myeloid cells, suggesting a cell-specific microglial inflammatory response in DIO. Conclusions: An obesogenic diet affects microglial immunometabolism in a time-of-day dependent manner. Given the central role of the brain in energy metabolism, a better knowledge of daily rhythms in microglial immunometabolism could lead to a better understanding of the pathogenesis of obesity.
Collapse
Affiliation(s)
- Irina V. Milanova
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Laboratory of Endocrinology, Amsterdam University Medical Center, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, Amsterdam, Netherlands
| | - Martin J. T. Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Laboratory of Endocrinology, Amsterdam University Medical Center, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, Amsterdam, Netherlands
| | - Xiao-Lan Wang
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Laboratory of Endocrinology, Amsterdam University Medical Center, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, Amsterdam, Netherlands
| | - Nikita L. Korpel
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Laboratory of Endocrinology, Amsterdam University Medical Center, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, Amsterdam, Netherlands
| | - Dirk Jan Stenvers
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Laboratory of Endocrinology, Amsterdam University Medical Center, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, Amsterdam, Netherlands
| | - Samantha E. C. Wolff
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Laboratory of Endocrinology, Amsterdam University Medical Center, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, Amsterdam, Netherlands
| | - Paul de Goede
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Laboratory of Endocrinology, Amsterdam University Medical Center, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, Amsterdam, Netherlands
| | - Annemieke C. Heijboer
- Laboratory of Endocrinology, Amsterdam University Medical Center, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, Amsterdam, Netherlands
- Endocrine Laboratory, Department of Clinical Chemistry, Amsterdam University Medical Center, Amsterdam Gastroenterology & Metabolism, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Laboratory of Endocrinology, Amsterdam University Medical Center, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, Amsterdam, Netherlands
| | - Susanne E. la Fleur
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Laboratory of Endocrinology, Amsterdam University Medical Center, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, Amsterdam, Netherlands
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Laboratory of Endocrinology, Amsterdam University Medical Center, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, Amsterdam, Netherlands
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Laboratory of Endocrinology, Amsterdam University Medical Center, Amsterdam Gastroenterology & Metabolism, University of Amsterdam, Amsterdam, Netherlands
- *Correspondence: Chun-Xia Yi
| |
Collapse
|
78
|
A comparison of hippocampal microglial responses in aged and young rodents following dependent and non-dependent binge drinking. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 148:305-343. [PMID: 31733666 PMCID: PMC9875180 DOI: 10.1016/bs.irn.2019.10.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Alcoholism is a highly visible and prevalent issue in the United States. Although binge-drinking is assumed to be a college-age problem, older adults (ages 65+) consume binge amounts of alcohol and have alcohol use disorders (AUDs). Moreover, individuals with alcohol dependence in their youth often continue to drink as they age. As such, this study tested the hypothesis that the effects of alcohol on hippocampal microglia are exacerbated in aged versus younger rodents in two AUD models. Briefly, adult (2-3 months) and aged (15+ months) Sprague-Dawley rats were administered alcohol or control diet using the Majchrowicz model to study alcohol-induced neurodegeneration. To study the effects of non-dependent binge consumption on microglia, adolescent (6-8 weeks) and aged (18+ months) C57/BL6N were subjected to the Drinking in the Dark paradigm. Microglia number and densitometry were assessed using immunohistochemistry. Hippocampal subregional and model/species-specific effects of alcohol were observed, but overall, aging did not appear to increase the alcohol-induced microglia reactivity as measured by Iba-1 densitometry. However, analysis of microglial counts revealed a significant decrease in the number microglia cells in both the alcohol-induced neurodegeneration and DID model across age groups. In the dentate gyrus, the loss of microglia was exacerbated by aging, particularly in mice after DID, non-dependent model. Using qRT-PCR, the persistence of alcohol and aging effects was assessed following the DID model. Allograft Inflammatory Factor 1 mRNA was increased in both young and aged mice by alcohol exposure; however, only in the aged mice did the alcohol effect persist. Overall, these data imply that the microglial response to alcohol is complex with evidence of depressed numbers of microglia but also increased reactivity with advanced age.
Collapse
|
79
|
Mendes NF, Kim YB, Velloso LA, Araújo EP. Hypothalamic Microglial Activation in Obesity: A Mini-Review. Front Neurosci 2018; 12:846. [PMID: 30524228 PMCID: PMC6262396 DOI: 10.3389/fnins.2018.00846] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 10/29/2018] [Indexed: 01/22/2023] Open
Abstract
Emerging data demonstrate that microglia activation plays a pivotal role in the development of hypothalamic inflammation in obesity. Early after the introduction of a high-fat diet, hypothalamic microglia undergo morphological, and functional changes in response to excessive dietary saturated fats. Initially the resident microglia are affected; however, as diet-induced obesity persists, bone marrow-derived myeloid cells gradually replace resident microglia. Genetic and pharmacological approaches aimed at dampening the inflammatory activity in the hypothalamus of experimental models of obesity have proven beneficial to correct the obese phenotype and improve metabolic abnormalities commonly associated with obesity. These approaches provide an experimental proof-of-concept that hypothalamic inflammation is central to the pathophysiology of obesity; understanding the details of the roles played by microglia in this process may help the development of preventive and therapeutic advances in the field. In this review, we discuss the potential mechanisms underlying hypothalamic microglial activation in high-fat induced obesity.
Collapse
Affiliation(s)
- Natália F Mendes
- School of Nursing, State University of Campinas, Campinas, Brazil.,Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism - Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.,Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
| | - Young-Bum Kim
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism - Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Lício A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation, Rio de Janeiro, Brazil
| | - Eliana P Araújo
- School of Nursing, State University of Campinas, Campinas, Brazil.,Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
| |
Collapse
|
80
|
Cork SC. The role of the vagus nerve in appetite control: Implications for the pathogenesis of obesity. J Neuroendocrinol 2018; 30:e12643. [PMID: 30203877 DOI: 10.1111/jne.12643] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 09/05/2018] [Accepted: 09/05/2018] [Indexed: 12/14/2022]
Abstract
The communication between the gut and the brain is important for the control of energy homeostasis. In response to food intake, enteroendocrine cells secrete gut hormones, which ultimately suppress appetite through centrally-mediated processes. Increasing evidence implicates the vagus nerve as an important conduit in transmitting these signals from the gastrointestinal tract to the brain. Studies have demonstrated that many of the gut hormones secreted from enteroendocrine cells signal through the vagus nerve, and the sensitivity of the vagus to these signals is regulated by feeding status. Furthermore, evidence suggests that a reduction in the ability of the vagus nerve to respond to the switch between a "fasted" and "fed" state, retaining sensitivity to orexigenic signals when fed or a reduced ability to respond to satiety hormones, may contribute to obesity. This review draws together the evidence that the vagus nerve is a crucial component of appetite regulation via the gut-brain axis, with a particular emphasis on experimental techniques and future developments.
Collapse
Affiliation(s)
- Simon C Cork
- Section of Endocrinology and Investigative Medicine, Division of Endocrinology, Diabetes and Metabolism, Imperial College London, London, UK
| |
Collapse
|
81
|
Oliveira ADAB, Melo NDFM, Vieira ÉDS, Nogueira PAS, Coope A, Velloso LA, Dezonne RS, Ueira-Vieira C, Botelho FV, Gomes JDAS, Zanon RG. Palmitate treated-astrocyte conditioned medium contains increased glutathione and interferes in hypothalamic synaptic network in vitro. Neurochem Int 2018; 120:140-148. [PMID: 30138641 DOI: 10.1016/j.neuint.2018.08.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 07/30/2018] [Accepted: 08/16/2018] [Indexed: 01/03/2023]
|
82
|
Abstract
The hypothalamus is the brain region responsible for the maintenance of energetic homeostasis. The regulation of this process arises from the ability of the hypothalamus to orchestrate complex physiological responses such as food intake and energy expenditure, circadian rhythm, stress response, and fertility. Metabolic alterations such as obesity can compromise these hypothalamic regulatory functions. Alterations in circadian rhythm, stress response, and fertility further contribute to aggravate the metabolic dysfunction of obesity and contribute to the development of chronic disorders such as depression and infertility.At cellular level, obesity caused by overnutrition can damage the hypothalamus promoting inflammation and impairing hypothalamic neurogenesis. Furthermore, hypothalamic neurons suffer apoptosis and impairment in synaptic plasticity that can compromise the proper functioning of the hypothalamus. Several factors contribute to these phenomena such as ER stress, oxidative stress, and impairments in autophagy. All these observations occur at the same time and it is still difficult to discern whether inflammatory processes are the main drivers of these cellular dysfunctions or if the hypothalamic hormone resistance (insulin, leptin, and ghrelin) can be pinpointed as the source of several of these events.Understanding the mechanisms that underlie the pathophysiology of obesity in the hypothalamus is crucial for the development of strategies that can prevent or attenuate the deleterious effects of obesity.
Collapse
|
83
|
Dalby MJ, Aviello G, Ross AW, Walker AW, Barrett P, Morgan PJ. Diet induced obesity is independent of metabolic endotoxemia and TLR4 signalling, but markedly increases hypothalamic expression of the acute phase protein, SerpinA3N. Sci Rep 2018; 8:15648. [PMID: 30353127 PMCID: PMC6199263 DOI: 10.1038/s41598-018-33928-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 10/08/2018] [Indexed: 01/17/2023] Open
Abstract
Hypothalamic inflammation is thought to contribute to obesity. One potential mechanism is via gut microbiota derived bacterial lipopolysaccharide (LPS) entering into the circulation and activation of Toll-like receptor-4. This is called metabolic endotoxemia. Another potential mechanism is systemic inflammation arising from sustained exposure to high-fat diet (HFD) over more than 12 weeks. In this study we show that mice fed HFD over 8 weeks become obese and show elevated plasma LPS binding protein, yet body weight gain and adiposity is not attenuated in mice lacking Tlr4 or its co-receptor Cd14. In addition, caecal microbiota composition remained unchanged by diet. Exposure of mice to HFD over a more prolonged period (20 weeks) to drive systemic inflammation also caused obesity. RNAseq used to assess hypothalamic inflammation in these mice showed increased hypothalamic expression of Serpina3n and Socs3 in response to HFD, with few other genes altered. In situ hybridisation confirmed increased Serpina3n and Socs3 expression in the ARC and DMH at 20-weeks, but also at 8-weeks and increased SerpinA3N protein could be detected as early as 1 week on HFD. Overall these data show lack of hypothalamic inflammation in response to HFD and that metabolic endotoxemia does not link HFD to obesity.
Collapse
Affiliation(s)
- Matthew J Dalby
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - Gabriella Aviello
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - Alexander W Ross
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - Alan W Walker
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - Perry Barrett
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - Peter J Morgan
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom.
| |
Collapse
|
84
|
Abstract
The regulation of energy and glucose balance contributes to whole-body metabolic homeostasis, and such metabolic regulation is disrupted in obesity and diabetes. Metabolic homeostasis is orchestrated partly in response to nutrient and vagal-dependent gut-initiated functions. Specifically, the sensory and motor fibres of the vagus nerve transmit intestinal signals to the central nervous system and exert biological and physiological responses. In the past decade, the understanding of the regulation of vagal afferent signals and of the associated metabolic effect on whole-body energy and glucose balance has progressed. This Review highlights the contributions made to the understanding of the vagal afferent system and examines the integrative role of the vagal afferent in gastrointestinal regulation of appetite and glucose homeostasis. Investigating the integrative and metabolic role of vagal afferent signalling represents a potential strategy to discover novel therapeutic targets to restore energy and glucose balance in diabetes and obesity.
Collapse
|
85
|
Chen W, Xia Z, Liu W, He X, Zhang W. Selective Vagotomy Worsens Glucose Control After Ileal Transposition. Obes Surg 2018; 28:2494-2499. [PMID: 29525935 DOI: 10.1007/s11695-018-3192-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
PURPOSES Our aim was to investigate the effects of selective celiac branch vagotomy on food intake and glycemic control after ileal transposition (IT) and the possible roles of the vagus on the improvement of diabetes. MATERIALS AND METHODS Forty non-obese rats with diabetes underwent either IT, IT + celiac branch vagotomy (ITV), sham IT (SI), or sham IT + celiac branch vagotomy (SIV). They were pair fed, and the food intake, body weight, fasting plasma glucose, and glucagon-like peptide 1 (GLP-1) level were monitored. The number of activated pro-opiomelanocortin (POMC) neurons and POMC-derived peptides were measured after sacrifice. RESULTS The fasting glucose level of the ITV group was higher (7.0 ± 0.7 mmol/L vs. 5.7 ± 0.3, P = 0.01), and the area under the curve of the oral glucose tolerance test (AUCOGTT) value was greater than that of the IT group (1101.8 ± 90.3 (mmol/l) min vs. 986.9 ± 47.7 (mmol/l) min, P = 0.01). There was no significant difference in the postprandial GLP-1 level between these two groups, but the number of activated neurons in the ITV group was less than that of the IT group (10.3 ± 2.1 vs. 14.9 ± 2.3, P < 0.01), while the relative content level of POMC-derived peptides in the ITV group was half that of the IT group (P < 0.01). CONCLUSIONS The celiac branches of the vagus might contribute to less eating and improvement of diabetes after IT. The activating vagus strategy might be a goal for the treatment of diabetes.
Collapse
Affiliation(s)
- Weijie Chen
- Department of Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Beijing, 100730, People's Republic of China
| | - Zenan Xia
- Department of Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Beijing, 100730, People's Republic of China
| | - Wei Liu
- Department of Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Beijing, 100730, People's Republic of China
| | - Xiaodong He
- Department of Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Beijing, 100730, People's Republic of China.
| | - Weimin Zhang
- Clinical Laboratory of Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1#, Beijing, 100730, People's Republic of China
| |
Collapse
|
86
|
Hypothalamic inflammation and malfunctioning glia in the pathophysiology of obesity and diabetes: Translational significance. Biochem Pharmacol 2018; 153:123-133. [PMID: 29337002 DOI: 10.1016/j.bcp.2018.01.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/09/2018] [Indexed: 12/25/2022]
|
87
|
Abstract
Portal hypertension is one cause and a part of a dynamic process triggered by chronic liver disease, mostly induced by alcohol or incorrect nutrition and less often by viral infections and autoimmune or genetic disease. Adequate staging - continuously modified by current knowledge - should guide the prevention and treatment of portal hypertension with defined endpoints. The main goals are interruption of etiology and prevention of complications followed, if necessary, by treatment of these. For the past few decades, shunts, mostly as intrahepatic stent bypass between portal and hepatic vein branches, have played an important role in the prevention of recurrent bleeding and ascites formation, although their impact on survival remains ambiguous. Systemic drugs, such as non-selective beta-blockers, statins, or antibiotics, reduce portal hypertension by decreasing intrahepatic resistance or portal tributary blood flow or by blunting inflammatory stimuli inside and outside the liver. Here, the interactions among the gut, liver, and brain are increasingly examined for new therapeutic options. There is no general panacea. The interruption of initiating factors is key. If not possible or if not possible in a timely manner, combined approaches should receive more attention before considering liver transplantation.
Collapse
Affiliation(s)
| | | | - Jonel Trebicka
- Department of Internal Medicine, University of Bonn, Bonn, Germany.,European Foundation for Study of Chronic Liver Failure, Barcelona, Spain
| |
Collapse
|
88
|
Clyburn C, Travagli RA, Browning KN. Acute high-fat diet upregulates glutamatergic signaling in the dorsal motor nucleus of the vagus. Am J Physiol Gastrointest Liver Physiol 2018; 314:G623-G634. [PMID: 29368945 PMCID: PMC6008060 DOI: 10.1152/ajpgi.00395.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Obesity is associated with dysregulation of vagal neurocircuits controlling gastric functions, including food intake and energy balance. In the short term, however, caloric intake is regulated homeostatically although the precise mechanisms responsible are unknown. The present study examined the effects of acute high-fat diet (HFD) on glutamatergic neurotransmission within central vagal neurocircuits and its effects on gastric motility. Sprague-Dawley rats were fed a control or HFD diet (14% or 60% kcal from fat, respectively) for 3-5 days. Whole cell patch-clamp recordings and brainstem application of antagonists were used to assess the effects of acute HFD on glutamatergic transmission to dorsal motor nucleus of the vagus (DMV) neurons and subsequent alterations in gastric tone and motility. After becoming hyperphagic initially, caloric balance was restored after 3 days following HFD exposure. In control rats, the non- N-methyl-d-aspartate (NMDA) receptor antagonist, 6,7-dinitroquinoxaline-2,3-dione (DNQX), but not the NMDA receptor antagonist, amino-5-phosphonopentanoate (AP5), significantly decreased excitatory synaptic currents and action potential firing rate in gastric-projecting DMV neurons. In contrast, both AP5 and DNQX decreased excitatory synaptic transmission and action potential firing in acute HFD neurons. When microinjected into the brainstem, AP5, but not DNQX, decreased gastric motility and tone in acute HFD rats only. These results suggest that acute HFD upregulates NMDA receptor-mediated currents, increasing DMV neuronal excitability and activating the vagal efferent cholinergic pathway, thus increasing gastric tone and motility. Although such neuroplasticity may be a persistent adaptation to the initial exposure to HFD, it may also be an important mechanism in homeostatic regulation of energy balance. NEW & NOTEWORTHY Vagal neurocircuits are critical to the regulation of gastric functions, including satiation and food intake. Acute high-fat diet upregulates glutamatergic signaling within central vagal neurocircuits via activation of N-methyl-d-aspartate receptors, increasing vagal efferent drive to the stomach. Although it is possible that such neuroplasticity is a persistent adaptation to initial exposure to the high-fat diet, it may also play a role in the homeostatic control of feeding.
Collapse
Affiliation(s)
- Courtney Clyburn
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - R. Alberto Travagli
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - Kirsteen N. Browning
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
89
|
Liao MJ, Lin H, He YW, Zou C. NFATc3 deficiency protects against high fat diet (HFD)-induced hypothalamus inflammation and apoptosis via p38 and JNK suppression. Biochem Biophys Res Commun 2018; 499:743-750. [PMID: 29596828 DOI: 10.1016/j.bbrc.2018.03.182] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 03/24/2018] [Indexed: 12/18/2022]
Abstract
Hypothalamic inflammation and apoptosis cause neural injury, playing an important role in metabolic syndrome development. Nuclear Factors of Activated T cells (NFATc3) show many physiological and pathological effects. However, the function of NFATc3 in high fat diet (HFD)-induced hypothalamus injury remains unknown. The wild type (WT) and NFATc3-knockout (KO) mice were subjected to HFD feeding for 16 weeks to examine NFATc3 function in vivo. Astrocytes isolated from WT or KO mice were cultured and exposed to fructose (Fru) in vitro. The liver damage, hypothalamus injury, pro-inflammatory markers, NF-κB (p65), Caspase-3 and mitogen-activated protein kinases (MAPKs) pathways were evaluated. NFATc3 was significantly up-regulated in hypothalamus from mice challenged with HFD, and in astrocytes incubated with Fru. Both in vivo and in vitro studies indicated that NFATc3-deletion attenuated metabolism syndrome, reduced inflammatory regulators expression, inactivated NF-κB (p65), Caspase-3 and p38/JNK signaling pathway. Of note, we identified that promoting p38 or JNK activation could rescue inflammatory response and apoptosis in NFATc3-KO astrocytes stimulated by Fru. Together, these findings revealed an important role of NFATc3 NFATc3 for HFD-induced metabolic syndrome and particularly hypothalamus injury, and understanding of the regulatory molecular mechanism might provide new and effective therapeutic strategies for prevention and treatment of hypothalamic damage associated with dietary obesity-associated neuroinflammation and apoptosis.
Collapse
Affiliation(s)
- Meng-Jun Liao
- Department of Anesthesiology, South China Hospital Affiliated to University of South China, Hengyang 421001, China
| | - Hua Lin
- Department of Anesthesia & surgery, BaoJi Municipal Central hospital, Baoji 721008, China
| | - Yun-Wu He
- Department of Pain, The Second Hospital Affiliated to University of South China, Hengyang 421001, China
| | - Cong Zou
- Department of Pain, The Second Hospital Affiliated to University of South China, Hengyang 421001, China.
| |
Collapse
|
90
|
Mulders RJ, de Git KCG, Schéle E, Dickson SL, Sanz Y, Adan RAH. Microbiota in obesity: interactions with enteroendocrine, immune and central nervous systems. Obes Rev 2018; 19:435-451. [PMID: 29363272 DOI: 10.1111/obr.12661] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/27/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023]
Abstract
Western diets, with high consumption of simple sugars and saturated fats, contribute to the rise in the prevalence of obesity. It now seems clear that high-fat diets cause obesity, at least in part, by modifying the composition and function of the microorganisms that colonize in the gastrointestinal tract, the microbiota. The exact pathways by which intestinal microbiota contribute to obesity remain largely unknown. High-fat diet-induced alterations in intestinal microbiota have been suggested to increase energy extraction, intestinal permeability and systemic inflammation while decreasing the capability to generate obesity-suppressing short-chain fatty acids. Moreover, by increasing systemic inflammation, microglial activation and affecting vagal nerve activity, 'obese microbiota' indirectly influence hypothalamic gene expression and promote overeating. Because the potential of intestinal microbiota to induce obesity has been recognized, multiple ways to modify its composition and function are being investigated to provide novel preventive and therapeutic strategies against diet-induced obesity.
Collapse
Affiliation(s)
- R J Mulders
- Master's Programme Science and Business Management, Utrecht University, Utrecht, The Netherlands
| | - K C G de Git
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - E Schéle
- Institute for Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - S L Dickson
- Institute for Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Y Sanz
- Microbial Ecology, Nutrition and Health Research Group, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - R A H Adan
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
91
|
Chen M, Qin X, Qiu L, Chen S, Zhou H, Xu Y, Hu Z, Zhang Y, Cao Q, Ying Z. Concentrated Ambient PM 2.5-Induced Inflammation and Endothelial Dysfunction in a Murine Model of Neural IKK2 Deficiency. ENVIRONMENTAL HEALTH PERSPECTIVES 2018; 126:027003. [PMID: 29410383 PMCID: PMC6066335 DOI: 10.1289/ehp2311] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 12/11/2017] [Accepted: 12/12/2017] [Indexed: 05/03/2023]
Abstract
BACKGROUND Exposure to ambient fine particulate matter (PM2.5) is associated with cardiovascular mortality, but underlying pathophysiologic mechanisms are not fully understood. Hypothalamic inflammation, characterized by the activation of Inhibitor kappaB kinase 2/Nuclear factor kappaB (IKK2/NF-κB) signaling pathway, may play an important role in the pathogenesis of cardiovascular diseases. We recently demonstrated that hypothalamic inflammation is increased in mice exposed to concentrated ambient PM2.5 (CAP). OBJECTIVES In the present study, we used a neuron-specific IKK2 knockout mouse model to examine the role of neural IKK2 expression and hypothalamic inflammation in the pathophysiologic effects of PM2.5. METHODS We assessed inflammatory and vascular responses in Nestin-creIKK2flox/flox (IKK2Neu-KO) and littermate Nestin-creIKK2flox/+ (control) mice after 4 mo of exposure to filtered air (FA) or CAP. RESULTS CAP exposure was associated with significantly higher tumor necrosis factor-α (TNFα) and interleukin (IL)-6 mRNA in the hypothalamus of control mice, but not IKK2Neu-KO mice. In addition, CAP exposure-induced increases in bronchoalveolar lavage fluid (BALF) leukocytes, pulmonary macrophage infiltration and IL-6 expression, plasma TNFα and IL-1β levels, adipose macrophage infiltration and IL-1β expression, and endothelial dysfunction were reduced or absent in IKK2Neu-KO mice compared with controls. CONCLUSIONS Our findings support a role of neural IKK2 in CAP exposure-induced local and systemic pro-inflammatory cytokine expression, pulmonary and adipose inflammation, and endothelial dysfunction, thus providing insight into pathophysiologic mechanisms that may mediate effects of PM2.5 exposure. https://doi.org/10.1289/EHP2311.
Collapse
Affiliation(s)
- Minjie Chen
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, China
| | - Xiaobo Qin
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Electrocardiography, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Lianglin Qiu
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Occupational and Environmental Health, School of Public Health, Nantong University, Nantong, China
| | - Sufang Chen
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Huifen Zhou
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Yanyi Xu
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, China
| | - Ziying Hu
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Endocrinology, The People's Hospital of Zhengzhou University (Henan Provincial People's Hospital) , Zhengzhou, Henan, China
| | - Yuhao Zhang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qi Cao
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Zhekang Ying
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, China
| |
Collapse
|
92
|
Naznin F, Toshinai K, Waise TMZ, Okada T, Sakoda H, Nakazato M. Restoration of metabolic inflammation-related ghrelin resistance by weight loss. J Mol Endocrinol 2018; 60:109-118. [PMID: 29233861 PMCID: PMC5793712 DOI: 10.1530/jme-17-0192] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 12/12/2017] [Indexed: 12/24/2022]
Abstract
High-fat diet (HFD)-induced metabolic inflammation in the central and peripheral organs contributes to the pathogenesis of obesity. Long-term HFD blunts signaling by ghrelin, a gastric-derived orexigenic peptide, in the vagal afferent nerve via a mechanism involving in situ activation of inflammation. This study was undertaken to investigate whether ghrelin resistance is associated with progressive development of metabolic inflammation. In mice, ghrelin's orexigenic activity was abolished 2-4 weeks after the commencement of HFD (60% of energy from fat), consistent with the timing of accumulation and activation of macrophages and microglia in the nodose ganglion and hypothalamus. Calorie-restricted weight loss after 12-week HFD feeding restored ghrelin responsiveness and alleviated the upregulation of macrophage/microglia activation markers and inflammatory cytokines. HSP72, a chaperone protein, was upregulated in the hypothalamus of HFD-fed mice, potentially contributing to prevention of irreversible neuron damage. These results demonstrate that ghrelin resistance is reversible following reversal of the HFD-induced inflammation and obesity phenotypes.
Collapse
Affiliation(s)
- Farhana Naznin
- Division of NeurologyRespirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Koji Toshinai
- Division of NeurologyRespirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Department of Sports and FitnessFaculty of Wellness, Shigakkan University, Obu, Japan
| | - T M Zaved Waise
- Division of NeurologyRespirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Tadashi Okada
- Division of NeurologyRespirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Hideyuki Sakoda
- Division of NeurologyRespirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Masamitsu Nakazato
- Division of NeurologyRespirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Agency for Medical Research and Development-Core Research for Evolutional Medical Science and Technology (AMED-CREST)Japan Agency for Medical Research and Development, Tokyo, Japan
| |
Collapse
|
93
|
The effects of overnight nutrient intake on hypothalamic inflammation in a free-choice diet-induced obesity rat model. Appetite 2018; 120:527-535. [PMID: 28988760 DOI: 10.1016/j.appet.2017.10.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/03/2017] [Accepted: 10/03/2017] [Indexed: 12/31/2022]
|
94
|
Woo YS, Bahk WM. The Link Between Obesity and Depression: Exploring Shared Mechanisms. UNDERSTANDING DEPRESSION 2018:203-220. [DOI: 10.1007/978-981-10-6577-4_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
95
|
Krustrup P, Helge EW, Hansen PR, Aagaard P, Hagman M, Randers MB, de Sousa M, Mohr M. Effects of recreational football on women's fitness and health: adaptations and mechanisms. Eur J Appl Physiol 2017; 118:11-32. [PMID: 29164325 DOI: 10.1007/s00421-017-3733-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/28/2017] [Indexed: 12/30/2022]
Abstract
The review describes the fitness and health effects of recreational football in women aged 18-65 years. The review documents that 2 × 1 h of recreational football training for 12-16 weeks causes marked improvements in maximal oxygen uptake (5-15%) and myocardial function in women. Moreover, mean arterial blood pressure was shown to decrease by 2-5 mmHg in normotensive women and 6-8 mmHg in hypertensive women. This review also show that short-term (< 4 months) and medium-term (4-16 months) recreational football training has major beneficial impact on metabolic health profile in women, with fat losses of 1-3 kg and improvements in blood lipid profile. Lastly, 2 × 1 h per week of recreational football training for women elevates lower extremity bone mineralisation by 1-5% and whole-body bone mineralization by 1-2% within 4-12-month interventions. These training adaptations are related to the high heart rates, high number of fast runs, and multiple changes of direction and speed occurring during recreational football training for untrained women. In conclusion, regular small-sided football training for women is an intense and versatile type of training that combines elements of high-intensity interval training (HIIT), endurance training and strength training, thereby providing optimal stimuli for cardiovascular, metabolic and musculoskeletal fitness. Recreational football, therefore, seems to be an effective tool for prevention and treatment of lifestyle diseases in young and middle-aged women, including hypertension, type 2 diabetes and osteopenia. Future research should elucidate effects of football training for elderly women, and as treatment and rehabilitation of breast cancer patients and other women patient groups.
Collapse
Affiliation(s)
- Peter Krustrup
- Department of Sports Science and Clinical Biomechanics, SDU Sport and Health Sciences Cluster (SHSC), Faculty of Health Sciences, University of Southern Denmark, Campusvej 55, 3450, Odense, Denmark. .,Sport and Health Sciences, College of Life and Environmental Sciences, St Luke's Campus, University of Exeter, Exeter, UK.
| | - Eva Wulff Helge
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Peter R Hansen
- Department of Cardiology, Herlev-Gentofte University Hospital, Gentofte, Denmark
| | - Per Aagaard
- Department of Sports Science and Clinical Biomechanics, SDU Sport and Health Sciences Cluster (SHSC), Faculty of Health Sciences, University of Southern Denmark, Campusvej 55, 3450, Odense, Denmark
| | - Marie Hagman
- Department of Sports Science and Clinical Biomechanics, SDU Sport and Health Sciences Cluster (SHSC), Faculty of Health Sciences, University of Southern Denmark, Campusvej 55, 3450, Odense, Denmark
| | - Morten B Randers
- Department of Sports Science and Clinical Biomechanics, SDU Sport and Health Sciences Cluster (SHSC), Faculty of Health Sciences, University of Southern Denmark, Campusvej 55, 3450, Odense, Denmark
| | - Maysa de Sousa
- Laboratory of Medical Investigation, LIM-18, Endocrinology Division, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Magni Mohr
- Department of Sports Science and Clinical Biomechanics, SDU Sport and Health Sciences Cluster (SHSC), Faculty of Health Sciences, University of Southern Denmark, Campusvej 55, 3450, Odense, Denmark.,Centre of Health Science, Faculty of Health Sciences, University of the Faroe Islands, Tórshavn, Faroe Islands.,Center of Health and Human Performance, Department of Food and Nutrition, and Sport Science, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
96
|
Abstract
The hypothalamus is an evolutionarily conserved brain structure that regulates an organism's basic functions, such as homeostasis and reproduction. Several hypothalamic nuclei and neuronal circuits have been the focus of many studies seeking to understand their role in regulating these basic functions. Within the hypothalamic neuronal populations, the arcuate melanocortin system plays a major role in controlling homeostatic functions. The arcuate pro-opiomelanocortin (POMC) neurons in particular have been shown to be critical regulators of metabolism and reproduction because of their projections to several brain areas both in and outside of the hypothalamus, such as autonomic regions of the brain stem and spinal cord. Here, we review and discuss the current understanding of POMC neurons from their development and intracellular regulators to their physiological functions and pathological dysregulation.
Collapse
Affiliation(s)
- Chitoku Toda
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Anna Santoro
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Jung Dae Kim
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Sabrina Diano
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520.,Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520.,Section of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
97
|
Insulin action and resistance in obesity and type 2 diabetes. Nat Med 2017; 23:804-814. [PMID: 28697184 DOI: 10.1038/nm.4350] [Citation(s) in RCA: 837] [Impact Index Per Article: 104.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 05/11/2017] [Indexed: 12/12/2022]
Abstract
Nutritional excess is a major forerunner of type 2 diabetes. It enhances the secretion of insulin, but attenuates insulin's metabolic actions in the liver, skeletal muscle and adipose tissue. However, conflicting evidence indicates a lack of knowledge of the timing of these events during the development of obesity and diabetes, pointing to a key gap in our understanding of metabolic disease. This Perspective reviews alternate viewpoints and recent results on the temporal and mechanistic connections between hyperinsulinemia, obesity and insulin resistance. Although much attention has addressed early steps in the insulin signaling cascade, insulin resistance in obesity seems to be largely elicited downstream of these steps. New findings also connect insulin resistance to extensive metabolic cross-talk between the liver, adipose tissue, pancreas and skeletal muscle. These and other advances over the past 5 years offer exciting opportunities and daunting challenges for the development of new therapeutic strategies for the treatment of type 2 diabetes.
Collapse
|
98
|
Guerville M, Leroy A, Sinquin A, Laugerette F, Michalski MC, Boudry G. Western-diet consumption induces alteration of barrier function mechanisms in the ileum that correlates with metabolic endotoxemia in rats. Am J Physiol Endocrinol Metab 2017; 313:E107-E120. [PMID: 28400412 DOI: 10.1152/ajpendo.00372.2016] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 04/03/2017] [Accepted: 04/04/2017] [Indexed: 12/11/2022]
Abstract
Obesity and its related disorders have been associated with the presence in the blood of gut bacteria-derived lipopolysaccharides (LPS). However, the factors underlying this low-grade elevation in plasma LPS, so-called metabolic endotoxemia, are not fully elucidated. We aimed to investigate the effects of Western diet (WD) feeding on intestinal and hepatic LPS handling mechanisms in a rat model of diet-induced obesity (DIO). Rats were fed either a standard chow diet (C) or a Western Diet (WD, 45% fat) for 6 wk. They were either fed ad libitum or pair-fed to match the caloric intake of C rats for the first week, then fed ad libitum for the remaining 5 wk. Six-week WD feeding led to a mild obese phenotype with increased adiposity and elevated serum LPS-binding protein (LBP) levels relative to C rats, irrespective of initial energy intake. Serum LPS was not different between dietary groups but exhibited strong variability. Disrupted ileal mucus secretion and decreased ileal Reg3-γ and -β gene expression along with high ileal permeability to LPS were observed in WD compared with C-fed rats. Ileal and cecal intestinal alkaline phosphatase (IAP) activity as well as Verrucomicrobia and Bifidobacterium cecal levels were increased in WD-fed rats compared with C-fed rats. WD consumption did not impact mRNA levels of LPS-handling hepatic enzymes. Correlation analysis revealed that ileal passage of LPS, IAP activity, Proteobacteria levels and hepatic aoah gene expression correlated with serum LPS and LBP, suggesting that ileal mucosal defense impairment induced by WD feeding contribute to metabolic endotoxemia.
Collapse
Affiliation(s)
- Mathilde Guerville
- Institut Numecan INRA INSERM Université de Rennes 1, Saint-Gilles, France; and
| | - Anaïs Leroy
- Institut Numecan INRA INSERM Université de Rennes 1, Saint-Gilles, France; and
| | - Annaëlle Sinquin
- Institut Numecan INRA INSERM Université de Rennes 1, Saint-Gilles, France; and
| | - Fabienne Laugerette
- Univ-Lyon, CarMeN Laboratory, INRA U1397, Université Claude Bernard Lyon 1, Inserm U1060, INSA Lyon, Villeurbanne, France
| | - Marie-Caroline Michalski
- Univ-Lyon, CarMeN Laboratory, INRA U1397, Université Claude Bernard Lyon 1, Inserm U1060, INSA Lyon, Villeurbanne, France
| | - Gaëlle Boudry
- Institut Numecan INRA INSERM Université de Rennes 1, Saint-Gilles, France; and
| |
Collapse
|
99
|
Miller GD. Appetite Regulation: Hormones, Peptides, and Neurotransmitters and Their Role in Obesity. Am J Lifestyle Med 2017; 13:586-601. [PMID: 31662725 DOI: 10.1177/1559827617716376] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/18/2017] [Accepted: 05/31/2017] [Indexed: 12/29/2022] Open
Abstract
Understanding body weight regulation will aid in the development of new strategies to combat obesity. This review examines energy homeostasis and food intake behaviors, specifically with regards to hormones, peptides, and neurotransmitters in the periphery and central nervous system, and their potential role in obesity. Dysfunction in feeding signals by the brain is a factor in obesity. The hypothalamic (arcuate nucleus) and brainstem (nucleus tractus solitaris) areas integrate behavioral, endocrine, and autonomic responses via afferent and efferent pathways from and to the brainstem and peripheral organs. Neurons present in the arcuate nucleus express pro-opiomelanocortin, Neuropeptide Y, and Agouti Related Peptide, with the former involved in lowering food intake, and the latter two acutely increasing feeding behaviors. Action of peripheral hormones from the gut, pancreas, adipose, and liver are also involved in energy homeostasis. Vagal afferent neurons are also important in regulating energy homeostasis. Peripheral signals respond to the level of stored and currently available fuel. By studying their actions, new agents maybe developed that disable orexigenic responses and enhance anorexigenic signals. Although there are relatively few medications currently available for obesity treatment, a number of agents are in development that work through these pathways.
Collapse
Affiliation(s)
- Gary D Miller
- Department of Health and Exercise Science, Wake Forest University, Winston-Salem, North Carolina
| |
Collapse
|
100
|
Vaughn AC, Cooper EM, DiLorenzo PM, O'Loughlin LJ, Konkel ME, Peters JH, Hajnal A, Sen T, Lee SH, de La Serre CB, Czaja K. Energy-dense diet triggers changes in gut microbiota, reorganization of gut‑brain vagal communication and increases body fat accumulation. Acta Neurobiol Exp (Wars) 2017; 77:18-30. [PMID: 28379213 DOI: 10.21307/ane-2017-033] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Obesity is associated with consumption of energy-dense diets and development of systemic inflammation. Gut microbiota play a role in energy harvest and inflammation and can influence the change from lean to obese phenotypes. The nucleus of the solitary tract (NTS) is a brain target for gastrointestinal signals modulating satiety and alterations in gut-brain vagal pathway may promote overeating and obesity. Therefore, we tested the hypothesis that high-fat diet‑induced changes in gut microbiota alter vagal gut-brain communication associated with increased body fat accumulation. Sprague-Dawley rats consumed a low energy‑dense rodent diet (LFD; 3.1 kcal/g) or high energy‑dense diet (HFD, 5.24 kcal/g). Minocycline was used to manipulate gut microbiota composition. 16S Sequencing was used to determine microbiota composition. Immunofluorescence against IB4 and Iba1 was used to determine NTS reorganization and microglia activation. Nodose ganglia from LFD rats were isolated and co-cultured with different bacteria strains to determine neurotoxicity. HFD altered gut microbiota with increases in Firmicutes/Bacteriodetes ratio and in pro-inflammatory Proteobacteria proliferation. HFD triggered reorganization of vagal afferents and microglia activation in the NTS, associated with weight gain. Minocycline-treated HFD rats exhibited microbiota profile comparable to LFD animals. Minocycline suppressed HFD‑induced reorganization of vagal afferents and microglia activation in the NTS, and reduced body fat accumulation. Proteobacteria isolated from cecum of HFD rats were toxic to vagal afferent neurons in culture. Our findings show that diet‑induced shift in gut microbiome may disrupt vagal gut‑brain communication resulting in microglia activation and increased body fat accumulation.
Collapse
Affiliation(s)
- Alexandra C Vaughn
- Washington State University, Integrative Physiology and Neuroscience, Pullman, WA, USA
| | - Erin M Cooper
- Washington State University, Integrative Physiology and Neuroscience, Pullman, WA, USA
| | | | - Levi J O'Loughlin
- Washington State University, School of Molecular Biosciences, Pullman, WA, USA
| | - Michael E Konkel
- Washington State University, School of Molecular Biosciences, Pullman, WA, USA
| | - James H Peters
- Washington State University, Integrative Physiology and Neuroscience, Pullman, WA, USA
| | - Andras Hajnal
- The Pennsylvania State University, College of Medicine, Neural and Behavioral Sciences, Hershey, PA, USA
| | - Tanusree Sen
- University of Georgia, Veterinary Biosciences and Diagnostic Imaging, Athens, GA, USA
| | - Sun Hye Lee
- University of Georgia, Foods and Nutrition, Athens, GA, USA
| | | | - Krzysztof Czaja
- University of Georgia, Veterinary Biosciences and Diagnostic Imaging, Athens, GA, USA,
| |
Collapse
|