1
|
Dali R, Langlet F. Tanycytes in the nexus of hypothalamic inflammation, appetite control, and obesity. Physiol Behav 2025; 296:114917. [PMID: 40222438 DOI: 10.1016/j.physbeh.2025.114917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/16/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
Hypothalamic inflammation has been identified as a critical factor driving the development of obesity and associated metabolic disorders. This inflammation-related disruption of energy balance relies on alterations in metabolic cues sensing and hypothalamic cellular functions, together leading to overeating and weight gain. Within the hypothalamic cellular networks controlling energy balance, recent studies have highlighted the significance of glial dysfunction in these processes, suggesting that these cells could provide new avenues for weight loss therapies. Glia rapidly activates following the consumption of a high-fat diet, even after a very short exposure, and contributes to the disruption of the entire system through inflammatory crosstalk. This review explores recent progress in understanding the molecular interactions between glial cells and neurons in hypothalamic inflammation related to obesity, diabetes, and associated complications. Notably, it highlights specialized ependymal cells called tanycytes, whose role is still underestimated in hypothalamic inflammation, and examines the potential for targeting this cell type as a treatment strategy for metabolic disorders.
Collapse
Affiliation(s)
- Rafik Dali
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland; Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Fanny Langlet
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland; Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
2
|
Yang K, Wu YT, He Y, Dai JX, Luo YL, Xie JH, Ding WJ. GLP-1 and IL-6 regulates obesity in the gut and brain. Life Sci 2025; 362:123339. [PMID: 39730038 DOI: 10.1016/j.lfs.2024.123339] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/06/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Obesity is a chronic metabolic disease characterized by excessive nutrient intake leading to increased subcutaneous or visceral fat, resulting in pathological and physiological changes. The incidence rate of obesity, an important form of metabolic syndrome, is increasing worldwide. Excess appetite is a key pathogenesis of obesity, and the inflammatory response induced by obesity has received increasing attention. This review focuses on the role of appetite-regulating factor (Glucogan-like peptide 1) and inflammatory factor (Interleukin-6) in the gut and brain in individuals with obesity and draws insights from the current literature.
Collapse
Affiliation(s)
- Kun Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yu-Ting Wu
- Chengdu University of Traditional Chinese Medicine, 1166 Luitai Avenue, Chengdu, Sichuan 611137, China
| | - Yan He
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jin-Xiu Dai
- Chengdu University of Traditional Chinese Medicine, 1166 Luitai Avenue, Chengdu, Sichuan 611137, China
| | - Yu-Lu Luo
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jing-Hui Xie
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Wei-Jun Ding
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
3
|
Díaz-Castro F, Morselli E, Claret M. Interplay between the brain and adipose tissue: a metabolic conversation. EMBO Rep 2024; 25:5277-5293. [PMID: 39558137 PMCID: PMC11624209 DOI: 10.1038/s44319-024-00321-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/20/2024] Open
Abstract
The central nervous system and adipose tissue interact through complex communication. This bidirectional signaling regulates metabolic functions. The hypothalamus, a key homeostatic brain region, integrates exteroceptive and interoceptive signals to control appetite, energy expenditure, glucose, and lipid metabolism. This regulation is partly achieved via the nervous modulation of white (WAT) and brown (BAT) adipose tissue. In this review, we highlight the roles of sympathetic and parasympathetic innervation in regulating WAT and BAT activities, such as lipolysis and thermogenesis. Adipose tissue, in turn, plays a dual role as an energy reservoir and an endocrine organ, secreting hormones that influence brain function and metabolic health. In addition, this review focuses on recently uncovered communication pathways, including extracellular vesicles and neuro-mesenchymal units, which add new layers of regulation and complexity to the brain-adipose tissue interaction. Finally, we also examine the consequences of disrupted communication between the brain and adipose tissue in metabolic disorders like obesity and type-2 diabetes, emphasizing the potential for new therapeutic strategies targeting these pathways to improve metabolic health.
Collapse
Affiliation(s)
- Francisco Díaz-Castro
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Laboratory of Autophagy and Metabolism, Faculty of Medicine and Sciences, Department of Basic Sciences, Universidad San Sebastián, Santiago de Chile, Chile
- Physiology Department, Biological Science Faculty, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
| | - Eugenia Morselli
- Laboratory of Autophagy and Metabolism, Faculty of Medicine and Sciences, Department of Basic Sciences, Universidad San Sebastián, Santiago de Chile, Chile.
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- IBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain.
- School of Medicine, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
4
|
Yamashima T. 4-Hydroxynonenal from Mitochondrial and Dietary Sources Causes Lysosomal Cell Death for Lifestyle-Related Diseases. Nutrients 2024; 16:4171. [PMID: 39683565 DOI: 10.3390/nu16234171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Excessive consumption of vegetable oils such as soybean and canolla oils containing ω-6 polyunsaturated fatty acids is considered one of the most important epidemiological factors leading to the progression of lifestyle-related diseases. However, the underlying mechanism of vegetable-oil-induced organ damage is incompletely elucidated. Since proopiomelanocortin (POMC) neurons in the hypothalamus are related to the control of appetite and energy expenditure, their cell degeneration/death is crucial for the occurrence of obesity. In patients with metabolic syndrome, saturated fatty acids, especially palmitate, are used as an energy source. Since abundant reactive oxygen species are produced during β-oxidation of the palmitate in mitochondria, an increased amount of 4-hydroxy-2-nonenal (4-HNE) is endogenously generated from linoleic acids constituting cardiolipin of the inner membranes. Further, due to the daily intake of deep-fried foods and/or high-fat diets cooked using vegetable oils, exogenous 4-HNE being generated via lipid peroxidation during heating is incorporated into the blood. By binding with atheromatous and/or senile plaques, 4-HNE inactivates proteins via forming hybrid covalent chemical addition compounds and causes cellular dysfunction and tissue damage by the specific oxidation carbonylation. 4-HNE overstimulates G-protein-coupled receptors to induce abnormal Ca2+ mobilization and µ-calpain activation. This endogenous and exogenous 4-HNE synergically causes POMC neuronal degeneration/death and obesity. Then, the resultant metabolic disorder facilitates degeneration/death of hippocampal neurons, pancreatic β-cells, and hepatocytes. Hsp70.1 is a molecular chaperone which is crucial for both protein quality control and the stabilization of lysosomal limiting membranes. Focusing on the monkey hippocampus after ischemia, previously we formulated the 'calpain-cathepsin hypothesis', i.e., that calpain-mediated cleavage of carbonylated Hsp70.1 is a trigger of programmed neuronal death. This review aims to report that in diverse organs, lysosomal cell degeneration/death occurs via the calpain-cathepsin cascade after the consecutive injections of synthetic 4-HNE in monkeys. Presumably, 4-HNE is a root substance of lysosomal cell death for lifestyle-related diseases.
Collapse
Affiliation(s)
- Tetsumori Yamashima
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Takara-machi 13-1, Kanazawa 920-8040, Japan
| |
Collapse
|
5
|
Bombassaro B, Araujo EP, Velloso LA. The hypothalamus as the central regulator of energy balance and its impact on current and future obesity treatments. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e240082. [PMID: 39876968 PMCID: PMC11771753 DOI: 10.20945/2359-4292-2024-0082] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/10/2024] [Indexed: 01/31/2025]
Abstract
The hypothalamus is a master regulator of energy balance in the body. First-order hypothalamic neurons localized in the arcuate nucleus sense systemic signals that indicate the energy stores in the body. Through distinct projections, arcuate nucleus neurons communicate with second-order neurons, which are mostly localized in the paraventricular nucleus and in the lateral hypothalamus. The signals then proceed to third- and fourth-order neurons that activate complex responses aimed at maintaining whole-body energy homeostasis. During the last 30 years, since the identification of leptin in 1994, there has been a great advance in the unveiling of the hypothalamic and extra-hypothalamic neuronal networks that control energy balance. This has contributed to the characterization of the mechanisms by which glucagon-like peptide-1 receptor agonists promote body mass reduction and has opened new windows of opportunity for the development of drugs to treat obesity. This review presents an overview of the mechanisms involved in the hypothalamic regulation of energy balance and discusses how advancements in this field are contributing to the development of new pharmacological strategies to treat obesity.
Collapse
Affiliation(s)
- Bruna Bombassaro
- Universidade de Campinas Centro de Pesquisa em Obesidade e Comorbidades CampinasSP Brasil Centro de Pesquisa em Obesidade e Comorbidades, Universidade de Campinas, Campinas, SP, Brasil
| | - Eliana P Araujo
- Universidade de Campinas Centro de Pesquisa em Obesidade e Comorbidades CampinasSP Brasil Centro de Pesquisa em Obesidade e Comorbidades, Universidade de Campinas, Campinas, SP, Brasil
| | - Licio A Velloso
- Universidade de Campinas Centro de Pesquisa em Obesidade e Comorbidades CampinasSP Brasil Centro de Pesquisa em Obesidade e Comorbidades, Universidade de Campinas, Campinas, SP, Brasil
| |
Collapse
|
6
|
Zhang Y, Wang R, Liu T, Wang R. Exercise as a Therapeutic Strategy for Obesity: Central and Peripheral Mechanisms. Metabolites 2024; 14:589. [PMID: 39590824 PMCID: PMC11596326 DOI: 10.3390/metabo14110589] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Obesity is a complex, multifactorial condition involving excessive fat accumulation due to an imbalance between energy intake and expenditure, with its global prevalence steadily rising. This condition significantly increases the risk of chronic diseases, including sarcopenia, type 2 diabetes, and cardiovascular diseases, highlighting the need for effective interventions. Exercise has emerged as a potent non-pharmacological approach to combat obesity, targeting both central and peripheral mechanisms that regulate metabolism, energy expenditure, and neurological functions. In the central nervous system, exercise influences appetite, mood, and cognitive functions by modulating the reward system and regulating appetite-controlling hormones to manage energy intake. Concurrently, exercise promotes thermogenesis in adipose tissue and regulates endocrine path-ways and key metabolic organs, such as skeletal muscle and the liver, to enhance fat oxidation and support energy balance. Despite advances in understanding exercise's role in obesity, the precise interaction between the neurobiological and peripheral metabolic pathways remains underexplored, particularly in public health strategies. A better understanding of these interactions could inform more comprehensive obesity management approaches by addressing both central nervous system influences on behavior and peripheral metabolic regulation. This review synthesizes recent insights into these roles, highlighting potential therapeutic strategies targeting both systems for more effective obesity interventions.
Collapse
Affiliation(s)
- Yiyin Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (Y.Z.); (R.W.)
| | - Ruwen Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (Y.Z.); (R.W.)
| | - Tiemin Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (Y.Z.); (R.W.)
| |
Collapse
|
7
|
Sánchez-Garrido MA, Serrano-López V, Ruiz-Pino F, Vázquez MJ, Rodríguez-Martín A, Torres E, Velasco I, Rodríguez AB, Chicano-Gálvez E, Mora-Ortiz M, Ohlsson C, Poutanen M, Pinilla L, Gaytán F, Douros JD, Yang B, Müller TD, DiMarchi RD, Tschöp MH, Finan B, Tena-Sempere M. Superior metabolic improvement of polycystic ovary syndrome traits after GLP1-based multi-agonist therapy. Nat Commun 2024; 15:8498. [PMID: 39353946 PMCID: PMC11445520 DOI: 10.1038/s41467-024-52898-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 09/25/2024] [Indexed: 10/03/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous condition, defined by oligo-/anovulation, hyper-androgenism and/or polycystic ovaries. Metabolic complications are common in patients suffering PCOS, including obesity, insulin resistance and type-2 diabetes, which severely compromise the clinical course of affected women. Yet, therapeutic options remain mostly symptomatic and of limited efficacy for the metabolic and reproductive alterations of PCOS. We report here the hormonal, metabolic and gonadal responses to the glucagon-like peptide-1 (GLP1)-based multi-agonists, GLP1/Estrogen (GLP1/E), GLP1/gastric inhibitory peptide (GLP1/GIP) and GLP1/GIP/Glucagon, in two mouse PCOS models, with variable penetrance of metabolic and reproductive traits, and their comparison with metformin. Our data illustrate the superior efficacy of GLP1/E vs. other multi-agonists and metformin in the management of metabolic complications of PCOS; GLP1/E ameliorates also ovarian cyclicity in an ovulatory model of PCOS, without direct estrogenic uterotrophic effects. In keeping with GLP1-mediated brain targeting, quantitative proteomics reveals changes in common and distinct hypothalamic pathways in response to GLP1/E between the two PCOS models, as basis for differential efficiency. Altogether, our data set the basis for the use of GLP1-based multi-agonists, and particularly GLP1/E, in the personalized management of PCOS.
Collapse
Affiliation(s)
- Miguel A Sánchez-Garrido
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain.
- Hospital Universitario Reina Sofía, Córdoba, Spain.
| | - Víctor Serrano-López
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Francisco Ruiz-Pino
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - María Jesús Vázquez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Andrea Rodríguez-Martín
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
| | - Encarnación Torres
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
| | - Inmaculada Velasco
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
| | - Ana Belén Rodríguez
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Córdoba, Spain
| | | | - Marina Mora-Ortiz
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
- Lipids & Atherosclerosis Unit, Reina Sofía University Hospital, Córdoba, Spain
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Matti Poutanen
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Leonor Pinilla
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Córdoba, Spain
| | - Francisco Gaytán
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Córdoba, Spain
| | | | - Bin Yang
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
- Walther-Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany
| | | | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technical University of München, Munich, Germany
| | - Brian Finan
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain.
- Hospital Universitario Reina Sofía, Córdoba, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Córdoba, Spain.
| |
Collapse
|
8
|
Ramadhan Marviano F, Santoso P, Rahayu R. Neuroprotective Effect of Colocasia esculenta Var. Mentawai Corm Flour High-Fat Diet Fed Mice. JOURNAL OF CELLULAR NEUROSCIENCE AND OXIDATIVE STRESS 2024; 16:1173-1182. [DOI: 10.37212/jcnos.1470198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
The transformation of our era, resulting in a change in dietary habits towards a higher intake of fatty foods, presents a worldwide health issue. Among these challenges is neurodegeneration, which leads to cognitive impairment. It is imperative to seek alternative solutions rooted in nature to address the limitations associated with non-natural treatment methods. This entails harnessing the properties of secondary metabolite compounds found in plants, such as Colocasia esculenta Var. Mentawai. This research aims to assess the efficacy of C. esculenta Var. Mentawai corm as neuroprotective agents in mitigating CNS damage and preventing cognitive decline associated with neurodegeneration. Daily administration of a high-fat diet and a mixture of taro flour is conducted on young adult male mice for a duration of 60 days. Furthermore, analysis of the neurocognitive ability of mice, determination of malondialdehyde levels, and observation of histopathological structures on brain tissue were carried out. The results showed that the group of mice fed with taro flour mixture effectively showed a positive impact on maintaining neurocognitive abilities and histopathological structure of brain tissue against neurodegeneration (p
Collapse
|
9
|
Le Thuc O, García-Cáceres C. Obesity-induced inflammation: connecting the periphery to the brain. Nat Metab 2024; 6:1237-1252. [PMID: 38997442 DOI: 10.1038/s42255-024-01079-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 06/11/2024] [Indexed: 07/14/2024]
Abstract
Obesity is often associated with a chronic, low-grade inflammatory state affecting the entire body. This sustained inflammatory state disrupts the coordinated communication between the periphery and the brain, which has a crucial role in maintaining homeostasis through humoural, nutrient-mediated, immune and nervous signalling pathways. The inflammatory changes induced by obesity specifically affect communication interfaces, including the blood-brain barrier, glymphatic system and meninges. Consequently, brain areas near the third ventricle, including the hypothalamus and other cognition-relevant regions, become susceptible to impairments, resulting in energy homeostasis dysregulation and an elevated risk of cognitive impairments such as Alzheimer's disease and dementia. This Review explores the intricate communication between the brain and the periphery, highlighting the effect of obesity-induced inflammation on brain function.
Collapse
Affiliation(s)
- Ophélia Le Thuc
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Cristina García-Cáceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
10
|
Rapps K, Marco A, Pe’er-Nissan H, Kisliouk T, Stemp G, Yadid G, Weller A, Meiri N. Exercise Rescues Obesogenic-Related Genes in the Female Hypothalamic Arcuate Nucleus: A Potential Role of miR-211 Modulation. Int J Mol Sci 2024; 25:7188. [PMID: 39000297 PMCID: PMC11241292 DOI: 10.3390/ijms25137188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Obesity is a major public health concern that is associated with negative health outcomes. Exercise and dietary restriction are commonly recommended to prevent or combat obesity. This study investigates how voluntary exercise mitigates abnormal gene expression in the hypothalamic arcuate nucleus (ARC) of diet-induced obese (DIO) rats. Using a transcriptomic approach, novel genes in the ARC affected by voluntary wheel running were assessed alongside physiology, pharmacology, and bioinformatics analysis to evaluate the role of miR-211 in reversing obesity. Exercise curbed weight gain and fat mass, and restored ARC gene expression. High-fat diet (HFD) consumption can dysregulate satiety/hunger mechanisms in the ARC. Transcriptional clusters revealed that running altered gene expression patterns, including inflammation and cellular structure genes. To uncover regulatory mechanisms governing gene expression in DIO attenuation, we explored miR-211, which is implicated in systemic inflammation. Exercise ameliorated DIO overexpression of miR-211, demonstrating its pivotal role in regulating inflammation in the ARC. Further, in vivo central administration of miR-211-mimic affected the expression of immunity and cell cycle-related genes. By cross-referencing exercise-affected and miR-211-regulated genes, potential candidates for obesity reduction through exercise were identified. This research suggests that exercise may rescue obesity through gene expression changes mediated partially through miR-211.
Collapse
Affiliation(s)
- Kayla Rapps
- Faculty of Life Sciences, Bar Ilan University, Ramat Gan 5290002, Israel; (K.R.); (H.P.-N.); (G.Y.)
- Institute of Animal Science, Agricultural Research Organization, The Volcani Center, Rishon LeZion 7528809, Israel;
- Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat Gan 5290002, Israel; (G.S.); (A.W.)
| | - Asaf Marco
- Neuro-Epigenetics Laboratory, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel;
| | - Hilla Pe’er-Nissan
- Faculty of Life Sciences, Bar Ilan University, Ramat Gan 5290002, Israel; (K.R.); (H.P.-N.); (G.Y.)
- Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat Gan 5290002, Israel; (G.S.); (A.W.)
| | - Tatiana Kisliouk
- Institute of Animal Science, Agricultural Research Organization, The Volcani Center, Rishon LeZion 7528809, Israel;
| | - Gabrielle Stemp
- Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat Gan 5290002, Israel; (G.S.); (A.W.)
| | - Gal Yadid
- Faculty of Life Sciences, Bar Ilan University, Ramat Gan 5290002, Israel; (K.R.); (H.P.-N.); (G.Y.)
- Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat Gan 5290002, Israel; (G.S.); (A.W.)
| | - Aron Weller
- Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat Gan 5290002, Israel; (G.S.); (A.W.)
- Department of Psychology, Bar Ilan University, Ramat Gan 5290002, Israel
| | - Noam Meiri
- Institute of Animal Science, Agricultural Research Organization, The Volcani Center, Rishon LeZion 7528809, Israel;
| |
Collapse
|
11
|
Manzo R, Gallardo-Becerra L, Díaz de León-Guerrero S, Villaseñor T, Cornejo-Granados F, Salazar-León J, Ochoa-Leyva A, Pedraza-Alva G, Pérez-Martínez L. Environmental Enrichment Prevents Gut Dysbiosis Progression and Enhances Glucose Metabolism in High-Fat Diet-Induced Obese Mice. Int J Mol Sci 2024; 25:6904. [PMID: 39000013 PMCID: PMC11241766 DOI: 10.3390/ijms25136904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/01/2024] [Accepted: 06/10/2024] [Indexed: 07/14/2024] Open
Abstract
Obesity is a global health concern implicated in numerous chronic degenerative diseases, including type 2 diabetes, dyslipidemia, and neurodegenerative disorders. It is characterized by chronic low-grade inflammation, gut microbiota dysbiosis, insulin resistance, glucose intolerance, and lipid metabolism disturbances. Here, we investigated the therapeutic potential of environmental enrichment (EE) to prevent the progression of gut dysbiosis in mice with high-fat diet (HFD)-induced metabolic syndrome. C57BL/6 male mice with obesity and metabolic syndrome, continuously fed with an HFD, were exposed to EE. We analyzed the gut microbiota of the mice by sequencing the 16s rRNA gene at different intervals, including on day 0 and 12 and 24 weeks after EE exposure. Fasting glucose levels, glucose tolerance, insulin resistance, food intake, weight gain, lipid profile, hepatic steatosis, and inflammatory mediators were evaluated in serum, adipose tissue, and the colon. We demonstrate that EE intervention prevents the progression of HFD-induced dysbiosis, reducing taxa associated with metabolic syndrome (Tepidimicrobium, Acidaminobacteraceae, and Fusibacter) while promoting those linked to healthy physiology (Syntrophococcus sucrumutans, Dehalobacterium, Prevotella, and Butyricimonas). Furthermore, EE enhances intestinal barrier integrity, increases mucin-producing goblet cell population, and upregulates Muc2 expression in the colon. These alterations correlate with reduced systemic lipopolysaccharide levels and attenuated colon inflammation, resulting in normalized glucose metabolism, diminished adipose tissue inflammation, reduced liver steatosis, improved lipid profiles, and a significant reduction in body weight gain despite mice's continued HFD consumption. Our findings highlight EE as a promising anti-inflammatory strategy for managing obesity-related metabolic dysregulation and suggest its potential in developing probiotics targeting EE-modulated microbial taxa.
Collapse
Affiliation(s)
- Rubiceli Manzo
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico
| | - Luigui Gallardo-Becerra
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico
| | - Sol Díaz de León-Guerrero
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico
| | - Tomas Villaseñor
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico
| | - Fernanda Cornejo-Granados
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico
| | - Jonathan Salazar-León
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico
| | - Adrian Ochoa-Leyva
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico
| | - Gustavo Pedraza-Alva
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico
| | - Leonor Pérez-Martínez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, Morelos, Mexico
| |
Collapse
|
12
|
Regina-Ferreira L, Valdivieso-Rivera F, Angelim MKSC, Menezes Dos Reis L, Furino VO, Morari J, Maia de Sousa L, Consonni SR, Sponton CH, Moraes-Vieira PM, Velloso LA. Inhibition of Crif1 protects fatty acid-induced POMC neuron-like cell-line damage by increasing CPT-1 function. Am J Physiol Endocrinol Metab 2024; 326:E681-E695. [PMID: 38597829 DOI: 10.1152/ajpendo.00420.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/07/2024] [Accepted: 03/27/2024] [Indexed: 04/11/2024]
Abstract
Hypothalamic proopiomelanocortin (POMC) neurons are sensors of signals that reflect the energy stored in the body. Inducing mild stress in proopiomelanocortin neurons protects them from the damage promoted by the consumption of a high-fat diet, mitigating the development of obesity; however, the cellular mechanisms behind these effects are unknown. Here, we induced mild stress in a proopiomelanocortin neuron cell line by inhibiting Crif1. In proopiomelanocortin neurons exposed to high levels of palmitate, the partial inhibition of Crif1 reverted the defects in mitochondrial respiration and ATP production; this was accompanied by improved mitochondrial fusion/fission cycling. Furthermore, the partial inhibition of Crif1 resulted in increased reactive oxygen species production, increased fatty acid oxidation, and reduced dependency on glucose for mitochondrial respiration. These changes were dependent on the activity of CPT-1. Thus, we identified a CPT-1-dependent metabolic shift toward greater utilization of fatty acids as substrates for respiration as the mechanism behind the protective effect of mild stress against palmitate-induced damage of proopiomelanocortin neurons.NEW & NOTEWORTHY Saturated fats can damage hypothalamic neurons resulting in positive energy balance, and this is mitigated by mild cellular stress; however, the mechanisms behind this protective effect are unknown. Using a proopiomelanocortin cell line, we show that under exposure to a high concentration of palmitate, the partial inhibition of the mitochondrial protein Crif1 results in protection due to a metabolic shift warranted by the increased expression and activity of the mitochondrial fatty acid transporter CPT-1.
Collapse
Affiliation(s)
| | - Fernando Valdivieso-Rivera
- Obesity and Comorbidities Research Center, São Paulo, Brazil
- Department of Structural and Functional Biology, Institute of Biology (IB), University of Campinas, São Paulo, Brazil
| | - Monara K S C Angelim
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, São Paulo, Brazil
| | - Larissa Menezes Dos Reis
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, São Paulo, Brazil
| | | | - Joseane Morari
- Obesity and Comorbidities Research Center, São Paulo, Brazil
| | - Lizandra Maia de Sousa
- Laboratory of Cytochemistry and Immunocytochemistry, Department of Biochemistry and Tissue Biology, Institute of Biology (IB), University of Campinas, São Paulo, Brazil
| | - Sílvio Roberto Consonni
- Laboratory of Cytochemistry and Immunocytochemistry, Department of Biochemistry and Tissue Biology, Institute of Biology (IB), University of Campinas, São Paulo, Brazil
| | - Carlos H Sponton
- Obesity and Comorbidities Research Center, São Paulo, Brazil
- Department of Structural and Functional Biology, Institute of Biology (IB), University of Campinas, São Paulo, Brazil
| | - Pedro M Moraes-Vieira
- Obesity and Comorbidities Research Center, São Paulo, Brazil
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, São Paulo, Brazil
| | - Lício A Velloso
- Obesity and Comorbidities Research Center, São Paulo, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation, São Paulo, Brazil
| |
Collapse
|
13
|
Leon S, Simon V, Lee TH, Steuernagel L, Clark S, Biglari N, Lesté-Lasserre T, Dupuy N, Cannich A, Bellocchio L, Zizzari P, Allard C, Gonzales D, Le Feuvre Y, Lhuillier E, Brochard A, Nicolas JC, Teillon J, Nikolski M, Marsicano G, Fioramonti X, Brüning JC, Cota D, Quarta C. Single cell tracing of Pomc neurons reveals recruitment of 'Ghost' subtypes with atypical identity in a mouse model of obesity. Nat Commun 2024; 15:3443. [PMID: 38658557 PMCID: PMC11043070 DOI: 10.1038/s41467-024-47877-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
The hypothalamus contains a remarkable diversity of neurons that orchestrate behavioural and metabolic outputs in a highly plastic manner. Neuronal diversity is key to enabling hypothalamic functions and, according to the neuroscience dogma, it is predetermined during embryonic life. Here, by combining lineage tracing of hypothalamic pro-opiomelanocortin (Pomc) neurons with single-cell profiling approaches in adult male mice, we uncovered subpopulations of 'Ghost' neurons endowed with atypical molecular and functional identity. Compared to 'classical' Pomc neurons, Ghost neurons exhibit negligible Pomc expression and are 'invisible' to available neuroanatomical approaches and promoter-based reporter mice for studying Pomc biology. Ghost neuron numbers augment in diet-induced obese mice, independent of neurogenesis or cell death, but weight loss can reverse this shift. Our work challenges the notion of fixed, developmentally programmed neuronal identities in the mature hypothalamus and highlight the ability of specialised neurons to reversibly adapt their functional identity to adult-onset obesogenic stimuli.
Collapse
Affiliation(s)
- Stéphane Leon
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Vincent Simon
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Thomas H Lee
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Lukas Steuernagel
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Samantha Clark
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Nasim Biglari
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | | | - Nathalie Dupuy
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Astrid Cannich
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Luigi Bellocchio
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Philippe Zizzari
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Camille Allard
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Delphine Gonzales
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Yves Le Feuvre
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Emeline Lhuillier
- University of Toulouse III Paul Sabatier, INSERM, Institut des Maladies Métaboliques et Cardiovasculaires, U1297, 31400, France; GeT-Santé, Plateforme Génome et Transcriptome, GenoToul, Toulouse, France
| | - Alexandre Brochard
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Jean Charles Nicolas
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Jérémie Teillon
- University of Bordeaux, CNRS, INSERM, BIC, US4, UAR 3420, F-33000, Bordeaux, France
| | - Macha Nikolski
- University of Bordeaux, Bordeaux Bioinformatics Center, Bordeaux, France
- University of Bordeaux, CNRS, IBGC UMR 5095, Bordeaux, France
| | - Giovanni Marsicano
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Xavier Fioramonti
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- National Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Daniela Cota
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Carmelo Quarta
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France.
| |
Collapse
|
14
|
Huang Y, Wang A, Zhou W, Li B, Zhang L, Rudolf AM, Jin Z, Hambly C, Wang G, Speakman JR. Maternal dietary fat during lactation shapes single nucleus transcriptomic profile of postnatal offspring hypothalamus in a sexually dimorphic manner in mice. Nat Commun 2024; 15:2382. [PMID: 38493217 PMCID: PMC10944494 DOI: 10.1038/s41467-024-46589-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 03/01/2024] [Indexed: 03/18/2024] Open
Abstract
Maternal overnutrition during lactation predisposes offspring to develop metabolic diseases and exacerbates the relevant syndromes in males more than females in later life. The hypothalamus is a heterogenous brain region that regulates energy balance. Here we combined metabolic trait quantification of mother and offspring mice under low and high fat diet (HFD) feeding during lactation, with single nucleus transcriptomic profiling of their offspring hypothalamus at peak lacation to understand the cellular and molecular alterations in response to maternal dietary pertubation. We found significant expansion in neuronal subpopulations including histaminergic (Hdc), arginine vasopressin/retinoic acid receptor-related orphan receptor β (Avp/Rorb) and agouti-related peptide/neuropeptide Y (AgRP/Npy) in male offspring when their mothers were fed HFD, and increased Npy-astrocyte interactions in offspring responding to maternal overnutrition. Our study provides a comprehensive offspring hypothalamus map at the peak lactation and reveals how the cellular subpopulations respond to maternal dietary fat in a sex-specific manner during development.
Collapse
Affiliation(s)
- Yi Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- Broad Institute of MIT and Harvard, Metabolism Program, Cambridge, MA, 02142, USA
| | - Anyongqi Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Wenjiang Zhou
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Centre for Evolutionary Biology, Fudan University, Shanghai, 200438, China
| | - Baoguo Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Linshan Zhang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Centre for Evolutionary Biology, Fudan University, Shanghai, 200438, China
| | - Agata M Rudolf
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zengguang Jin
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Catherine Hambly
- School of Biological Sciences, University of Aberdeen, Aberdeen, AB24 3FX, UK
| | - Guanlin Wang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Centre for Evolutionary Biology, Fudan University, Shanghai, 200438, China.
| | - John R Speakman
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- School of Biological Sciences, University of Aberdeen, Aberdeen, AB24 3FX, UK.
- China Medical University, Shenyang, Liaoning, 110122, China.
| |
Collapse
|
15
|
Rossi C, Distaso M, Raggi F, Kusmic C, Faita F, Solini A. Lacking P2X7-receptors protects substantia nigra dopaminergic neurons and hippocampal-related cognitive performance from the deleterious effects of high-fat diet exposure in adult male mice. Front Nutr 2024; 11:1289750. [PMID: 38344021 PMCID: PMC10854005 DOI: 10.3389/fnut.2024.1289750] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/11/2024] [Indexed: 01/05/2025] Open
Abstract
BACKGROUND Dietary fat consumption, involved in the pathogenesis of insulin resistance and impaired glucose metabolism, is linked with decline in cognitive functions, dementia, and development of Parkinson's disease and Alzheimer's disease. Mature IL-1β, requiring the activation of the P2X7 receptor (P2X7R)-inflammasome complex, is an important mediator of neuroinflammation. The aim of the study was to test whether P2X7R activation might interfere with systemic and cerebral metabolic homeostasis. METHODS We treated WT and P2X7R KO mice with a high-fat diet (HFD) for 16 weeks, evaluating the effects on the Substantia Nigra and Hippocampus, target areas of damage in several forms of cognitive impairment. RESULTS HFD-treated WT and P2X7R KO mice showed a different brain mRNA profile of Insulin and Igf-1, with these genes and relative receptors, more expressed in KO mice. Unlike P2X7R KO mice, WT mice treated with HFD displayed a diameter reduction in dopaminergic neurons in the Substantia Nigra, accompanied by an increased IBA1 expression in this area; they also showed poor performances during Y-Maze and Morris Water Maze, tasks involving Hippocampus activity. Conversely, Parkin, whose reduction might promote neuronal cell death, was increased in the brain of P2X7R KO animals. CONCLUSION We report for the first time that HFD induces damage in dopaminergic neurons of the Substantia Nigra and a Hippocampus-related worse cognitive performance, both attenuated in the absence of P2X7R. The involved mechanisms might differ in the two brain areas, with a predominant role of inflammation in the Substantia Nigra and a metabolic derangement in the Hippocampus.
Collapse
Affiliation(s)
- Chiara Rossi
- Department of Surgical, Medical, Molecular and Critical Area Pathology University of Pisa, Pisa, Italy
| | - Mariarosaria Distaso
- Department of Surgical, Medical, Molecular and Critical Area Pathology University of Pisa, Pisa, Italy
| | - Francesco Raggi
- Department of Surgical, Medical, Molecular and Critical Area Pathology University of Pisa, Pisa, Italy
| | | | | | - Anna Solini
- Department of Surgical, Medical, Molecular and Critical Area Pathology University of Pisa, Pisa, Italy
| |
Collapse
|
16
|
Lopes PKF, Costa SDO, Simino LADP, Chaves WF, Silva FA, Costa CL, Milanski M, Ignacio-Souza LM, Torsoni AS, Torsoni MA. Hypothalamic inflammation and the development of an obese phenotype induced by high-fat diet consumption is exacerbated in alpha7 nicotinic cholinergic receptor knockout mice. Food Res Int 2024; 176:113808. [PMID: 38163714 DOI: 10.1016/j.foodres.2023.113808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/27/2023] [Accepted: 12/02/2023] [Indexed: 01/03/2024]
Abstract
Hypothalamic inflammation and metabolic changes resulting from the consumption of high-fat diets have been linked to low grade inflammation and obesity. Inflammation impairs the hypothalamic expression of α7 nicotinic acetylcholine receptor (α7nAChR). The α7nAChR is described as the main component of the anti-inflammatory cholinergic pathway in different inflammation models. To assess whether the reduction in α7nAChR expression exacerbates hypothalamic inflammation induced by a high-fat diet (HFD), were used male and female global α7nAChR knockout mouse line in normal or high-fat diet for 4 weeks. Body weight gain, adiposity, glucose homeostasis, hypothalamic inflammation, food intake, and energy expenditure were evaluated. Insulin sensitivity was evaluated in neuronal cell culture. Consumption of an HFD for 4 weeks resulted in body weight gain and adiposity in male Chrna7-/- mice and the hypothalamus of male Chrna7-/- mice showed neuroinflammatory markers, with increased gene expression of pro-inflammatory cytokines and dysregulation in the nuclear factor kappa B pathway. Moreover, male Chrna7-/- mice consuming an HFD showed alterations in glucose homeostasis and serum of Chrna7-/- mice that consumed an HFD impaired insulin signalling in neuronal cell culture experiments. In general, female Chrna7-/- mice that consumed an HFD did not show the phenotypic and molecular changes found in male mice, indicating that there is sexual dimorphism in the analysed parameters. Thus, receptor deletion resulted in increased susceptibility to hypothalamic inflammation and metabolic damage associated with HFD consumption in male mice.
Collapse
Affiliation(s)
| | - Suleyma de Oliveira Costa
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil
| | - Laís A de Paula Simino
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil
| | - Wenicios Ferreira Chaves
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil
| | - Franciely Alves Silva
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil
| | - Caroline Lobo Costa
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil
| | - Marciane Milanski
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil; Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Brazil
| | - Leticia Martins Ignacio-Souza
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil; Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Brazil
| | - Adriana Souza Torsoni
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil; Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Brazil
| | - Marcio Alberto Torsoni
- Laboratory of Metabolic Disorders (Labdime), School of Applied Sciences, University of Campinas (UNICAMP), Brazil; Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Brazil.
| |
Collapse
|
17
|
Jörgensen SK, Karnošová A, Mazzaferro S, Rowley O, Chen HJC, Robbins SJ, Christofides S, Merkle FT, Maletínská L, Petrik D. An analogue of the Prolactin Releasing Peptide reduces obesity and promotes adult neurogenesis. EMBO Rep 2024; 25:351-377. [PMID: 38177913 PMCID: PMC10897398 DOI: 10.1038/s44319-023-00016-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 11/02/2023] [Accepted: 11/17/2023] [Indexed: 01/06/2024] Open
Abstract
Hypothalamic Adult Neurogenesis (hAN) has been implicated in regulating energy homeostasis. Adult-generated neurons and adult Neural Stem Cells (aNSCs) in the hypothalamus control food intake and body weight. Conversely, diet-induced obesity (DIO) by high fat diets (HFD) exerts adverse influence on hAN. However, the effects of anti-obesity compounds on hAN are not known. To address this, we administered a lipidized analogue of an anti-obesity neuropeptide, Prolactin Releasing Peptide (PrRP), so-called LiPR, to mice. In the HFD context, LiPR rescued the survival of adult-born hypothalamic neurons and increased the number of aNSCs by reducing their activation. LiPR also rescued the reduction of immature hippocampal neurons and modulated calcium dynamics in iPSC-derived human neurons. In addition, some of these neurogenic effects were exerted by another anti-obesity compound, Liraglutide. These results show for the first time that anti-obesity neuropeptides influence adult neurogenesis and suggest that the neurogenic process can serve as a target of anti-obesity pharmacotherapy.
Collapse
Affiliation(s)
| | - Alena Karnošová
- First Faculty of Medicine, Charles University, Prague, 12108, Czech Republic
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, 16610, Czech Republic
| | - Simone Mazzaferro
- Wellcome-MRC Institute of Metabolic Science, Cambridge, CB2 0QQ, UK
- Wellcome-MRC Stem Cell Institute, Cambridge, CB2 0AW, UK
| | - Oliver Rowley
- School of Biosciences, Cardiff University, Cardiff, CF10 3AX, UK
| | - Hsiao-Jou Cortina Chen
- Wellcome-MRC Institute of Metabolic Science, Cambridge, CB2 0QQ, UK
- Wellcome-MRC Stem Cell Institute, Cambridge, CB2 0AW, UK
| | - Sarah J Robbins
- School of Biosciences, Cardiff University, Cardiff, CF10 3AX, UK
| | | | - Florian T Merkle
- Wellcome-MRC Institute of Metabolic Science, Cambridge, CB2 0QQ, UK
- Wellcome-MRC Stem Cell Institute, Cambridge, CB2 0AW, UK
| | - Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, 16610, Czech Republic
| | - David Petrik
- School of Biosciences, Cardiff University, Cardiff, CF10 3AX, UK.
| |
Collapse
|
18
|
Expression of Concern: High-Fat Diet Induces Apoptosis of Hypothalamic Neurons. PLoS One 2023; 18:e0292912. [PMID: 37816007 PMCID: PMC10564158 DOI: 10.1371/journal.pone.0292912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2023] Open
|
19
|
Sa M, Yoo ES, Koh W, Park MG, Jang HJ, Yang YR, Bhalla M, Lee JH, Lim J, Won W, Kwon J, Kwon JH, Seong Y, Kim B, An H, Lee SE, Park KD, Suh PG, Sohn JW, Lee CJ. Hypothalamic GABRA5-positive neurons control obesity via astrocytic GABA. Nat Metab 2023; 5:1506-1525. [PMID: 37653043 DOI: 10.1038/s42255-023-00877-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/25/2023] [Indexed: 09/02/2023]
Abstract
The lateral hypothalamic area (LHA) regulates food intake and energy balance. Although LHA neurons innervate adipose tissues, the identity of neurons that regulate fat is undefined. Here we show that GABRA5-positive neurons in LHA (GABRA5LHA) polysynaptically project to brown and white adipose tissues in the periphery. GABRA5LHA are a distinct subpopulation of GABAergic neurons and show decreased pacemaker firing in diet-induced obesity mouse models in males. Chemogenetic inhibition of GABRA5LHA suppresses fat thermogenesis and increases weight gain, whereas gene silencing of GABRA5 in LHA decreases weight gain. In the diet-induced obesity mouse model, GABRA5LHA are tonically inhibited by nearby reactive astrocytes releasing GABA, which is synthesized by monoamine oxidase B (Maob). Gene silencing of astrocytic Maob in LHA facilitates fat thermogenesis and reduces weight gain significantly without affecting food intake, which is recapitulated by administration of a Maob inhibitor, KDS2010. We propose that firing of GABRA5LHA suppresses fat accumulation and selective inhibition of astrocytic GABA is a molecular target for treating obesity.
Collapse
Affiliation(s)
- Moonsun Sa
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seongbuk-gu, Seoul, Republic of Korea
| | - Eun-Seon Yoo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Wuhyun Koh
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Mingu Gordon Park
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Hyun-Jun Jang
- Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Yong Ryoul Yang
- Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Mridula Bhalla
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- IBS School, University of Science and Technology, Daejeon, Republic of Korea
| | - Jae-Hun Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Jiwoon Lim
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- IBS School, University of Science and Technology, Daejeon, Republic of Korea
| | - Woojin Won
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Jea Kwon
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Joon-Ho Kwon
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Yejin Seong
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Byungeun Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, University of Science and Technology, Daejeon, Republic of Korea
| | - Heeyoung An
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Seung Eun Lee
- Virus Facility, Research Animal Resource Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Ki Duk Park
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, University of Science and Technology, Daejeon, Republic of Korea
| | - Pann-Ghill Suh
- Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Jong-Woo Sohn
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea.
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seongbuk-gu, Seoul, Republic of Korea.
- IBS School, University of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
20
|
Cargnin-Carvalho A, da Silva MR, Costa AB, Engel NA, Farias BX, Bressan JB, Backes KM, de Souza F, da Rosa N, de Oliveira Junior AN, Goldim MPDS, Correa MEAB, Venturini LM, Fortunato JJ, Prophiro JS, Petronilho F, Silveira PCL, Ferreira GK, Rezin GT. High concentrations of fructose cause brain damage in mice. Biochem Cell Biol 2023; 101:313-325. [PMID: 36947832 DOI: 10.1139/bcb-2022-0088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023] Open
Abstract
Excessive fructose consumption is associated with the incidence of obesity and systemic inflammation, resulting in increased oxidative damage and failure to the function of brain structures. Thus, we hypothesized that fructose consumption will significantly increase inflammation, oxidative damage, and mitochondrial dysfunction in the mouse brain and, consequently, memory damage. The effects of different fructose concentrations on inflammatory and biochemical parameters in the mouse brain were evaluated. Male Swiss mice were randomized into four groups: control, with exclusive water intake, 5%, 10%, and 20% fructose group. The 10% and 20% fructose groups showed an increase in epididymal fat, in addition to higher food consumption. Inflammatory markers were increased in epididymal fat and in some brain structures. In the evaluation of oxidative damage, it was possible to observe significant increases in the hypothalamus, prefrontal cortex, and hippocampus. In the epididymal fat and in the prefrontal cortex, there was a decrease in the activity of the mitochondrial respiratory chain complexes and an increase in the striatum. Furthermore, short memory was impaired in the 10% and 20% groups but not long memory. In conclusion, excess fructose consumption can cause fat accumulation, inflammation, oxidative damage, and mitochondrial dysfunction, which can damage brain structures and consequently memory.
Collapse
Affiliation(s)
- Anderson Cargnin-Carvalho
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Mariella Reinol da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Ana Beatriz Costa
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Nicole Alessandra Engel
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Bianca Xavier Farias
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Joice Benedet Bressan
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Kassiane Mathiola Backes
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Francielly de Souza
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Naiana da Rosa
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Aloir Neri de Oliveira Junior
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Mariana Pereira de Souza Goldim
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | | | - Ligia Milanez Venturini
- Laboratory of Experimental Phisiopatology, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Santa Catarina, Brazil
| | - Jucélia Jeremias Fortunato
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Josiane Somariva Prophiro
- Immunoparasitology Research Group, Postgraduate Program in Health Sciences, Universidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Fabrícia Petronilho
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| | - Paulo Cesar Lock Silveira
- Laboratory of Experimental Phisiopatology, Postgraduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Santa Catarina, Brazil
| | | | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health ScienceUniversidade do Sul de Santa Catarina, Santa Catarina, Brazil
| |
Collapse
|
21
|
Becetti I, Bwenyi EL, de Araujo IE, Ard J, Cryan JF, Farooqi IS, Ferrario CR, Gluck ME, Holsen LM, Kenny PJ, Lawson EA, Lowell BB, Schur EA, Stanley TL, Tavakkoli A, Grinspoon SK, Singhal V. The Neurobiology of Eating Behavior in Obesity: Mechanisms and Therapeutic Targets: A Report from the 23rd Annual Harvard Nutrition Obesity Symposium. Am J Clin Nutr 2023; 118:314-328. [PMID: 37149092 PMCID: PMC10375463 DOI: 10.1016/j.ajcnut.2023.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/03/2023] [Accepted: 05/01/2023] [Indexed: 05/08/2023] Open
Abstract
Obesity is increasing at an alarming rate. The effectiveness of currently available strategies for the treatment of obesity (including pharmacologic, surgical, and behavioral interventions) is limited. Understanding the neurobiology of appetite and the important drivers of energy intake (EI) can lead to the development of more effective strategies for the prevention and treatment of obesity. Appetite regulation is complex and is influenced by genetic, social, and environmental factors. It is intricately regulated by a complex interplay of endocrine, gastrointestinal, and neural systems. Hormonal and neural signals generated in response to the energy state of the organism and the quality of food eaten are communicated by paracrine, endocrine, and gastrointestinal signals to the nervous system. The central nervous system integrates homeostatic and hedonic signals to regulate appetite. Although there has been an enormous amount of research over many decades regarding the regulation of EI and body weight, research is only now yielding potentially effective treatment strategies for obesity. The purpose of this article is to summarize the key findings presented in June 2022 at the 23rd annual Harvard Nutrition Obesity Symposium entitled "The Neurobiology of Eating Behavior in Obesity: Mechanisms and Therapeutic Targets." Findings presented at the symposium, sponsored by NIH P30 Nutrition Obesity Research Center at Harvard, enhance our current understanding of appetite biology, including innovative techniques used to assess and systematically manipulate critical hedonic processes, which will shape future research and the development of therapeutics for obesity prevention and treatment.
Collapse
Affiliation(s)
- Imen Becetti
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States.
| | - Esther L Bwenyi
- Metabolism Unit, Massachusetts General Hospital, Boston, MA, United States; Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Ivan E de Araujo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY, United States; Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Jamy Ard
- Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Bariatric and Weight Management Center, Wake Forest Baptist Health, Winston-Salem, NC, United States; Center on Diabetes, Obesity, and Metabolism, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Hypertension and Vascular Research Center, Cardiovascular Sciences Center, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Maya Angelou Center for Healthy Equity, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Ismaa Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom; Wellcome-Medical Research Council (MRC) Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom; Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, United Kingdom
| | - Carrie R Ferrario
- Department of Pharmacology, Psychology Department (Biopsychology Area), University of Michigan, Ann Arbor, MI, United States
| | - Marci E Gluck
- National Institutes of Health, Phoenix, AZ, United States; National Institute of Diabetes and Digestive and Kidney Disease, Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, Phoenix, AZ, United States
| | - Laura M Holsen
- Harvard Medical School, Boston, MA, United States; Division of Women's Health, Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States; Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, United States
| | - Paul J Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY, United States; Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Elizabeth A Lawson
- Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States; Neuroendocrine Unit, Massachusetts General Hospital, Boston, MA, United States
| | - Bradford B Lowell
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Ellen A Schur
- Division of General Internal Medicine, University of Washington, Seattle, WA, United States; Univeristy of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, United States; Univeristy of Washington Nutrition and Obesity Research Center, University of Washington, Seattle, WA, United States; Clinical and Translational Research Services Core, University of Washington, Seattle, WA, United States
| | - Takara L Stanley
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States; Metabolism Unit, Massachusetts General Hospital, Boston, MA, United States; Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Ali Tavakkoli
- Division of General and Gastrointestinal (GI) Surgery, Center for Weight Management and Wellness, Advanced Minimally Invasive Fellowship, Harvard Medical School, Boston, MA, United States
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital, Boston, MA, United States; Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Vibha Singhal
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States; Harvard Medical School, Boston, MA, United States; Pediatric Endocrinology and Obesity Medicine, Massachusetts General Hospital, Boston, MA, United States; Pediatric Program MGH Weight Center, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
22
|
Lu C, Zhang J, Wang B, Gao Q, Ma K, Pei S, Li J, Cui S. Casein kinase 1α is required to maintain murine hypothalamic pro-opiomelanocortin expression. iScience 2023; 26:106670. [PMID: 37168577 PMCID: PMC10165255 DOI: 10.1016/j.isci.2023.106670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/08/2023] [Accepted: 04/11/2023] [Indexed: 05/13/2023] Open
Abstract
Hypothalamic pro-opiomelanocortin (POMC) neuron development is considered to play an essential role in the development of obesity. However, the underlying mechanisms remain unclear. Casein kinase 1α (CK1α) was expressed in the embryonic mouse hypothalamus at high levels and colocalized with POMC neurons. CK1α deletion in POMC neurons caused weight gain, metabolic defects, and increased food intake. The number of POMC-expressing cells was considerably decreased in Csnk1a1fl/fl;POMCcre (PKO) mice from embryonic day 15.5 to postnatal day 60, while apoptosis of POMC neurons was not affected. Furthermore, unchanged POMC progenitor cells and a decreased POMC phenotype established CK1α function in hypothalamic POMC neuron development. CK1α deletion led to elevated Notch intracellular domain (NICD) protein expression, and NICD inhibition rescued the PKO mouse phenotype. In summary, CK1α is involved in hypothalamic POMC expression via NICD-POMC signaling, deepening our understanding of POMC neuron development and control of systemic metabolic functions.
Collapse
Affiliation(s)
- Chenyang Lu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
| | - Jinglin Zhang
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
| | - Bingjie Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
| | - Qiao Gao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
| | - Kezhe Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
| | - Shaona Pei
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
| | - Juxue Li
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Sheng Cui
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, Jiangsu 225009, People’s Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, People’s Republic of China
- Corresponding author
| |
Collapse
|
23
|
Sewaybricker LE, Huang A, Chandrasekaran S, Melhorn SJ, Schur EA. The Significance of Hypothalamic Inflammation and Gliosis for the Pathogenesis of Obesity in Humans. Endocr Rev 2023; 44:281-296. [PMID: 36251886 DOI: 10.1210/endrev/bnac023] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/12/2022] [Indexed: 11/19/2022]
Abstract
Accumulated preclinical literature demonstrates that hypothalamic inflammation and gliosis are underlying causal components of diet-induced obesity in rodent models. This review summarizes and synthesizes available translational data to better understand the applicability of preclinical findings to human obesity and its comorbidities. The published literature in humans includes histopathologic analyses performed postmortem and in vivo neuroimaging studies measuring indirect markers of hypothalamic tissue microstructure. Both support the presence of hypothalamic inflammation and gliosis in children and adults with obesity. Findings predominantly point to tissue changes in the region of the arcuate nucleus of the hypothalamus, although findings of altered tissue characteristics in whole hypothalamus or other hypothalamic regions also emerged. Moreover, the severity of hypothalamic inflammation and gliosis has been related to comorbid conditions, including glucose intolerance, insulin resistance, type 2 diabetes, and low testosterone levels in men, independent of elevated body adiposity. Cross-sectional findings are augmented by a small number of prospective studies suggesting that a greater degree of hypothalamic inflammation and gliosis may predict adiposity gain and worsening insulin sensitivity in susceptible individuals. In conclusion, existing human studies corroborate a large preclinical literature demonstrating that hypothalamic neuroinflammatory responses play a role in obesity pathogenesis. Extensive or permanent hypothalamic tissue remodeling may negatively affect the function of neuroendocrine regulatory circuits and promote the development and maintenance of elevated body weight in obesity and/or comorbid endocrine disorders.
Collapse
Affiliation(s)
| | - Alyssa Huang
- Department of Pediatrics, University of Washington, Division of Endocrinology and Diabetes, Seattle Children's Hospital, Seattle, WA 98015, USA
| | | | - Susan J Melhorn
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Ellen A Schur
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
24
|
Jeczmien-Lazur JS, Sanetra AM, Pradel K, Izowit G, Chrobok L, Palus-Chramiec K, Piggins HD, Lewandowski MH. Metabolic cues impact non-oscillatory intergeniculate leaflet and ventral lateral geniculate nucleus: standard versus high-fat diet comparative study. J Physiol 2023; 601:979-1016. [PMID: 36661095 DOI: 10.1113/jp283757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 01/12/2023] [Indexed: 01/21/2023] Open
Abstract
The intergeniculate leaflet and ventral lateral geniculate nucleus (IGL/VLG) are subcortical structures involved in entrainment of the brain's circadian system to photic and non-photic (e.g. metabolic and arousal) cues. Both receive information about environmental light from photoreceptors, exhibit infra-slow oscillations (ISO) in vivo, and connect to the master circadian clock. Although current evidence demonstrates that the IGL/VLG communicate metabolic information and are crucial for entrainment of circadian rhythms to time-restricted feeding, their sensitivity to food intake-related peptides has not been investigated yet. We examined the effect of metabolically relevant peptides on the spontaneous activity of IGL/VLG neurons. Using ex vivo and in vivo electrophysiological recordings as well as in situ hybridisation, we tested potential sensitivity of the IGL/VLG to anorexigenic and orexigenic peptides, such as cholecystokinin, glucagon-like peptide 1, oxyntomodulin, peptide YY, orexin A and ghrelin. We explored neuronal responses to these drugs during day and night, and in standard vs. high-fat diet conditions. We found that IGL/VLG neurons responded to all the substances tested, except peptide YY. Moreover, more neurons responded to anorexigenic drugs at night, while a high-fat diet affected the IGL/VLG sensitivity to orexigenic peptides. Interestingly, ISO neurons responded to light and orexin A, but did not respond to the other food intake-related peptides. In contrast, non-ISO cells were activated by metabolic peptides, with only some being responsive to light. Our results show for the first time that peptides involved in the body's energy homeostasis stimulate the thalamus and suggest functional separation of the IGL/VLG cells. KEY POINTS: The intergeniculate leaflet and ventral lateral geniculate nucleus (IGL/VLG) of the rodent thalamus process various signals and participate in circadian entrainment. In both structures, cells exhibiting infra-slow oscillatory activity as well as non-rhythmically firing neurons being observed. Here, we reveal that only one of these two groups of cells responds to anorexigenic (cholecystokinin, glucagon-like peptide 1 and oxyntomodulin) and orexigenic (ghrelin and orexin A) peptides. Neuronal responses vary depending on the time of day (day vs. night) and on the diet (standard vs. high-fat diet). Additionally, we visualised receptors to the tested peptides in the IGL/VLG using in situ hybridisation. Our results suggest that two electrophysiologically different subpopulations of IGL/VLG neurons are involved in two separate functions: one related to the body's energy homeostasis and one associated with the subcortical visual system.
Collapse
Affiliation(s)
- Jagoda S Jeczmien-Lazur
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Anna M Sanetra
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Kamil Pradel
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Gabriela Izowit
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Lukasz Chrobok
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland.,School of Physiology, Pharmacology, and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - Katarzyna Palus-Chramiec
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Hugh D Piggins
- School of Physiology, Pharmacology, and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - Marian H Lewandowski
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| |
Collapse
|
25
|
Metabolic Activation of PARP as a SARS-CoV-2 Therapeutic Target-Is It a Bait for the Virus or the Best Deal We Could Ever Make with the Virus? Is AMBICA the Potential Cure? Biomolecules 2023; 13:biom13020374. [PMID: 36830743 PMCID: PMC9953159 DOI: 10.3390/biom13020374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/18/2023] Open
Abstract
The COVID-19 pandemic has had a great impact on global health and is an economic burden. Even with vaccines and anti-viral medications we are still scrambling to get a balance. In this perspective, we have shed light upon an extremely feasible approach by which we can control the SARS-CoV-2 infection and the associated complications, bringing some solace to this ongoing turmoil. We are providing some insights regarding an ideal agent which could prevent SARS-CoV-2 multiplication. If we could identify an agent which is an activator of metabolism and is also bioactive, we could prevent corona activation (AMBICA). Some naturally occurring lipid molecules best fit this identity as an agent which has the capacity to replenish our host cells, specifically immune cells, with ATP. It could also act as a source for providing a substrate for host cell PARP family members for MARylation and PARylation processes, leading to manipulation of the viral macro domain function, resulting in curbing the virulence and propagation of SARS-CoV-2. Identification of the right lipid molecule or combination of lipid molecules will fulfill the criteria. This perspective has focused on a unique angle of host-pathogen interaction and will open up a new dimension in treating COVID-19 infection.
Collapse
|
26
|
Migliolo L, de A. Boleti A, de O. Cardoso P, Frihling BF, e Silva P, de Moraes LRN. Adipose tissue, systematic inflammation, and neurodegenerative diseases. Neural Regen Res 2023; 18:38-46. [PMID: 35799506 PMCID: PMC9241402 DOI: 10.4103/1673-5374.343891] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
27
|
Griffin H, Sullivan SC, Barger SW, Phelan KD, Baldini G. Liraglutide Counteracts Endoplasmic Reticulum Stress in Palmitate-Treated Hypothalamic Neurons without Restoring Mitochondrial Homeostasis. Int J Mol Sci 2022; 24:ijms24010629. [PMID: 36614074 PMCID: PMC9820707 DOI: 10.3390/ijms24010629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/19/2022] [Accepted: 12/24/2022] [Indexed: 12/31/2022] Open
Abstract
One feature of high-fat diet-induced neurodegeneration in the hypothalamus is an increased level of palmitate, which is associated with endoplasmic reticulum (ER) stress, loss of CoxIV, mitochondrial fragmentation, and decreased abundance of MC4R. To determine whether antidiabetic drugs protect against ER and/or mitochondrial dysfunction by lipid stress, hypothalamic neurons derived from pre-adult mice and neuronal Neuro2A cells were exposed to elevated palmitate. In the hypothalamic neurons, palmitate exposure increased expression of ER resident proteins, including that of SERCA2, indicating ER stress. Liraglutide reverted such altered ER proteostasis, while metformin only normalized SERCA2 expression. In Neuro2A cells liraglutide, but not metformin, also blunted dilation of the ER induced by palmitate treatment, and enhanced abundance and expression of MC4R at the cell surface. Thus, liraglutide counteracts, more effectively than metformin, altered ER proteostasis, morphology, and folding capacity in neurons exposed to fat. In palmitate-treated hypothalamic neurons, mitochondrial fragmentation took place together with loss of CoxIV and decreased mitochondrial membrane potential (MMP). Metformin, but not liraglutide, reverted mitochondrial fragmentation, and both liraglutide and metformin did not protect against either loss of CoxIV abundance or MMP. Thus, ER recovery from lipid stress can take place in hypothalamic neurons in the absence of recovered mitochondrial homeostasis.
Collapse
Affiliation(s)
- Haven Griffin
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Sarah C. Sullivan
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Steven W. Barger
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Kevin D. Phelan
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Giulia Baldini
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Correspondence:
| |
Collapse
|
28
|
Natrus L, Osadchuk Y, Lisakovska O, Roch T, Babel N, Klys Y, Labudzynskyi D, Chaikovsky Y. Regulation of the apoptosis/autophagy switch by propionic acid in ventromedial hypothalamus of rats with type 2 diabetes mellitus. Heliyon 2022; 8:e11529. [PMID: 36439719 PMCID: PMC9681650 DOI: 10.1016/j.heliyon.2022.e11529] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 06/07/2022] [Accepted: 11/04/2022] [Indexed: 11/16/2022] Open
Abstract
Background Hypothalamic dysregulation may cause abnormal glucose metabolism and type 2 diabetes mellitus (T2DM). The balance between autophagy and apoptosis is important for maintaining cellular/tissue homeostasis and may be disrupted in T2DM. Objectives Since propionic acid (PA) exerts neuroprotective effects, the aim was to investigate its effects on apoptosis/autophagy switch in the ventromedial hypothalamus (VMH) of T2DM rats. Materials and methods Male Wistar rats were divided: 1) control; 2) T2DM; groups that received (14 days, orally): 3) metformin (60 mg/kg); 4) sodium salt of PA (60 mg/kg); 5) PA + metformin. Western blotting (Bax, Bcl-xl, LC3, Beclin-1, caspase-3), RT-PCR (Bax, Bcl-xl, LC3, Beclin-1), transmission electron microscopy and immunohistochemical staining (Bax, Bcl-xl) were performed on the VMH samples. Results T2DM-induced apoptosis and mitoptosis, enlarged endoplasmic reticulum (ER) tubules/cisterns were observed in VMH, and accompanied by an imbalance of pro- and anti-apoptotic factors: elevation of pro-apoptotic markers Bax and caspase-3, decrease in autophagy protein LC3 and anti-apoptotic Bcl-xl. Metformin and PA administration partially improved VMH ultrastructural changes by reducing mitochondrial swelling and diminishing the number of apoptotic neurons. Metformin inhibited neuronal apoptosis, however, caused reactive astrogliosis and accumulation of lipofuscin granules. Elevated number of autophagosomes was associated with the LC3, Beclin-1 and Bcl-xl increase and decrease in Bax and caspase-3 vs. T2DM. PA switched cell fate from apoptosis to autophagy by elevating LC3 and Beclin-1 levels, increasing Bcl-xl content that altogether may represent adaptive response to T2DM-induced apoptosis. PA + metformin administration lowered relative area of ER membranes/cisterns vs. control, T2DM and metformin, and was optimal considering ratio between the pro-apoptotic, anti-apoptotic and autophagy markers. Conclusion T2DM was associated with apoptosis activation leading to impairments in VMH. PA in combination with metformin may be effective against diabetes-induced cell death by switching apoptosis to autophagy in VMH.
Collapse
Affiliation(s)
- Larysa Natrus
- Department of Modern Technologies of Medical Diagnostics & Treatment, Bogomolets National Medical University, 34 Peremoha Avenue, Kyiv 03115, Ukraine
| | - Yuliia Osadchuk
- Department of Histology and Embryology, Bogomolets National Medical University, 34 Peremoha Avenue, Kyiv 03115, Ukraine
| | - Olha Lisakovska
- Department of Biochemistry of Vitamins and Coenzymes, Palladin Institute of Biochemistry, 9 Leontovicha Str., Kyiv 01054, Ukraine
| | - Toralf Roch
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr University Bochum, Hölkeskampring 40, 44625 Herne, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Center for Advanced Therapies (BeCAT), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Nina Babel
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr University Bochum, Hölkeskampring 40, 44625 Herne, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Center for Advanced Therapies (BeCAT), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Yuliia Klys
- Department of Modern Technologies of Medical Diagnostics & Treatment, Bogomolets National Medical University, 34 Peremoha Avenue, Kyiv 03115, Ukraine
| | - Dmytro Labudzynskyi
- Department of Biochemistry of Vitamins and Coenzymes, Palladin Institute of Biochemistry, 9 Leontovicha Str., Kyiv 01054, Ukraine
| | - Yuri Chaikovsky
- Department of Histology and Embryology, Bogomolets National Medical University, 34 Peremoha Avenue, Kyiv 03115, Ukraine
| |
Collapse
|
29
|
Li S, Luo X, Liao Z, Xu H, Liang M, Mai K, Zhang Y. Additional supplementation of sulfur-containing amino acids in the diets improves the intestinal health of turbot fed high-lipid diets. FISH & SHELLFISH IMMUNOLOGY 2022; 130:368-379. [PMID: 36115604 DOI: 10.1016/j.fsi.2022.09.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/29/2022] [Accepted: 09/10/2022] [Indexed: 06/15/2023]
Abstract
An eight-week feeding trial was conducted to investigate the effects of diets supplemented with three sulfur-containing amino acids (SAA), namely, methionine, cysteine, and taurine, on the intestinal health status of juvenile turbot (Scophthalmus maximus) fed high-lipid diets. Four diets were formulated, namely, a high-lipid control diet (16% lipid, HL) and three SAA-supplemented diets, which were formulated by supplementing 1.5% methionine (HLM), 1.5% cysteine (HLC), and 1.5% taurine (HLT) into the HL control diet, respectively. Each diet was assigned to triplicate tanks, and each tank was stocked with 30 juvenile fish (appr. initial weight, 8 g). The histological and morphometric results showed that dietary SAA supplementation obviously improved the intestinal morphology and integrity, in particular as reflected by higher height of microvilli and mucosal folds. Dietary SAA supplementation, in particular cysteine, up-regulated the gene expression of mucin-2 and tight junction proteins (ZO-1, Tricellilun and JAM). Dietary SAA supplementation remarkably down-regulated the gene expression of apoptosis-related factors such as p38, JNK, and Bax, expression of pro-inflammatory factors (e.g., NF-κB, AP-1 IL-1β, IL-8, and TNF-α). SAA supplementation resulted in higher antioxidative abilities in the intestine. Additionally, dietary SAA supplementation largely altered the communities of intestinal microbiota. Compared with the HL group, higher relative abundance of potential beneficial bacteria, and lower relative abundance of opportunistic pathogens were observed in SAA-supplemented groups. Dietary taurine supplementation significantly increased the relative abundance of Ligilactobacillus (in particular Lactobacillus murinus) and Limosilactobacillus (especially Lactobacillus reuteri). In conclusion, dietary sulfur-containing amino acids supplementation have promising potential in ameliorating the intestinal inflammation of turbot fed high-lipid diets. Especially dietary cysteine and taurine supplementation have more positive effects on the communities of the intestinal microbiota of turbot.
Collapse
Affiliation(s)
- Sihui Li
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Xing Luo
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China
| | - Zhangbin Liao
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China
| | - Houguo Xu
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China; Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, China.
| | - Mengqing Liang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China; Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China; Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, China
| | - Yanjiao Zhang
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China; Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, China.
| |
Collapse
|
30
|
Varela L, Horvath TL. Paraventricular glia drive circuit function to control metabolism. Cell Metab 2022; 34:1424-1426. [PMID: 36198288 DOI: 10.1016/j.cmet.2022.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The role of glia as active participants in brain functions has become increasingly evident. In this issue of Cell Metabolism, Herrera Moro Chao et al. reveal that astrocytes in the hypothalamic paraventricular nucleus (PVN) bidirectionally control neuronal behavior in response to metabolic cues and that this control is disrupted in obesity.
Collapse
Affiliation(s)
- Luis Varela
- Laboratory of Neuron-Glia Interaction in the Control of Hunger, Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain; IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Tamas L Horvath
- Laboratory of Neuron-Glia Interaction in the Control of Hunger, Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain; IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain; Department of Comparative Medicine and Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
31
|
Hydrogen Sulfide Attenuates High-Fat Diet-Induced Obesity: Involvement of mTOR/IKK/NF-κB Signaling Pathway. Mol Neurobiol 2022; 59:6903-6917. [PMID: 36053437 DOI: 10.1007/s12035-022-03004-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 08/16/2022] [Indexed: 10/14/2022]
Abstract
Obesity has become a public health epidemic worldwide and is associated with many diseases with high mortality including hypertension, diabetes, and heart disease. High-fat diet (HFD)-induced energy imbalance is one of the primary causes of obesity, but the underlying mechanisms are not fully elucidated. Our study showed that HFD reduced the level of hydrogen sulfide (H2S) and its catalytic enzyme cystathionine β-synthase (CBS) in mouse hypothalamus and plasma. We found that HFD activated mTOR, IKK/NF-κB, the main pathway regulating inflammation. Activation of inflammatory pathway promoted the production of pro-inflammatory cytokines including IL-6, IL-1β, and TNF-α, which caused cell damage and loss in the hypothalamus. The disturbance of the hypothalamic neuron circuits resulted in body weight gain in HFD-induced mice. Importantly, we also showed that restoration of H2S level with NaHS or activation of CBS with SAMe attenuated HFD-induced activation of mTOR, IKK/NF-κB signaling, which reduced the inflammation and the neuronal cell loss in the hypothalamus, and also inhibited body weight gain in mice. The same effects were obtained by inhibiting mTOR or NF-κB, which suggested that mTOR and NF-κB were the critical molecular factors involved in hypothalamic inflammation. Taken together, this study identified that HFD-induced hypothalamus inflammation plays a critical role in the development of obesity. Moreover, the inhibition of hypothalamic inflammation by regaining H2S level could be a potential therapeutic to prevent the development of obesity.
Collapse
|
32
|
Galyamina AG, Smagin DA, Kovalenko IL, Redina OE, Babenko VN, Kudryavtseva NN. The Dysfunction of Carcinogenesis- and Apoptosis-Associated Genes that Develops in the Hypothalamus under Chronic Social Defeat Stress in Male Mice. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:1050-1064. [PMID: 36180995 DOI: 10.1134/s0006297922090152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 08/25/2022] [Accepted: 08/25/2022] [Indexed: 06/16/2023]
Abstract
Chronic social stress caused by daily agonistic interactions in male mice leads to a mixed anxiety/depression-like disorder that is accompanied by the development of psychogenic immunodeficiency and stimulation of oncogenic processes concurrently with many neurotranscriptomic changes in brain regions. The aim of the study was to identify carcinogenesis- and apoptosis-associated differentially expressed genes (DEGs) in the hypothalamus of male mice with depression-like symptoms and, for comparison, in aggressive male mice with positive social experience. To obtain two groups of animals with the opposite 20-day social experiences, a model of chronic social conflict was used. Analysis of RNA-Seq data revealed similar expression changes for many DEGs between the aggressive and depressed animals in comparison with the control group; however, the number of DEGs was significantly lower in the aggressive than in the depressed mice. It is likely that the observed unidirectional changes in the expression of carcinogenesis- and apoptosis-associated genes in the two experimental groups may be a result of prolonged social stress (of different severity) caused by the agonistic interactions. In addition, 26 DEGs were found that did not change expression in the aggressive animals and could be considered genes promoting carcinogenesis or inhibiting apoptosis. Akt1, Bag6, Foxp4, Mapk3, Mapk8, Nol3, Pdcd10, and Xiap were identified as genes whose expression most strongly correlated with the expression of other DEGs, suggesting that their protein products play a role in coordination of the neurotranscriptomic changes in the hypothalamus. Further research into functions of these genes may be useful for the development of pharmacotherapies for psychosomatic pathologies.
Collapse
Affiliation(s)
- Anna G Galyamina
- FRC Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Dmitry A Smagin
- FRC Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Irina L Kovalenko
- FRC Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Olga E Redina
- FRC Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Vladimir N Babenko
- FRC Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Natalia N Kudryavtseva
- FRC Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia.
- Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, 199034, Russia
| |
Collapse
|
33
|
Fernández‐Arjona MDM, León‐Rodríguez A, Grondona JM, López‐Ávalos MD. Long-term priming of hypothalamic microglia is associated with energy balance disturbances under diet-induced obesity. Glia 2022; 70:1734-1761. [PMID: 35603807 PMCID: PMC9540536 DOI: 10.1002/glia.24217] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/13/2022] [Accepted: 05/06/2022] [Indexed: 12/16/2022]
Abstract
Exposure of microglia to an inflammatory environment may lead to their priming and exacerbated response to future inflammatory stimuli. Here we aimed to explore hypothalamic microglia priming and its consequences on energy balance regulation. A model of intracerebroventricular administration of neuraminidase (NA, which is present in various pathogens such as influenza virus) was used to induce acute neuroinflammation. Evidences of primed microglia were observed 3 months after NA injection, namely (1) a heightened response of microglia located in the hypothalamic arcuate nucleus after an in vivo inflammatory challenge (high fat diet [HFD] feeding for 10 days), and (2) an enhanced response of microglia isolated from NA-treated mice and challenged in vitro to LPS. On the other hand, the consequences of a previous NA-induced neuroinflammation were further evaluated in an alternative inflammatory and hypercaloric scenario, such as the obesity generated by continued HDF feeding. Compared with sham-injected mice, NA-treated mice showed increased food intake and, surprisingly, reduced body weight. Besides, NA-treated mice had enhanced microgliosis (evidenced by increased number and reactive morphology of microglia) and a reduced population of POMC neurons in the basal hypothalamus. Thus, a single acute neuroinflammatory event may elicit a sustained state of priming in microglial cells, and in particular those located in the hypothalamus, with consequences in hypothalamic cytoarchitecture and its regulatory function upon nutritional challenges.
Collapse
Affiliation(s)
- María del Mar Fernández‐Arjona
- Instituto de Investigación Biomédica de Málaga‐IBIMAMálagaSpain
- Grupo de investigación en Neuropsicofarmacología, Laboratorio de Medicina RegenerativaHospital Regional Universitario de MálagaMálagaSpain
| | - Ana León‐Rodríguez
- Instituto de Investigación Biomédica de Málaga‐IBIMAMálagaSpain
- Departamento de Biología Celular, Genética y Fisiología, Facultad de CienciasUniversidad de Málaga, Campus de TeatinosMálagaSpain
| | - Jesús M. Grondona
- Instituto de Investigación Biomédica de Málaga‐IBIMAMálagaSpain
- Departamento de Biología Celular, Genética y Fisiología, Facultad de CienciasUniversidad de Málaga, Campus de TeatinosMálagaSpain
| | - María D. López‐Ávalos
- Instituto de Investigación Biomédica de Málaga‐IBIMAMálagaSpain
- Departamento de Biología Celular, Genética y Fisiología, Facultad de CienciasUniversidad de Málaga, Campus de TeatinosMálagaSpain
| |
Collapse
|
34
|
TRIM67 Deficiency Exacerbates Hypothalamic Inflammation and Fat Accumulation in Obese Mice. Int J Mol Sci 2022; 23:ijms23169438. [PMID: 36012700 PMCID: PMC9409122 DOI: 10.3390/ijms23169438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/12/2022] [Accepted: 08/19/2022] [Indexed: 11/29/2022] Open
Abstract
Obesity has achieved the appearance of a global epidemic and is a serious cause for concern. The hypothalamus, as the central regulator of energy homeostasis, plays a critical role in regulating food intake and energy expenditure. In this study, we show that TRIM67 in the hypothalamus was responsive to body-energy homeostasis whilst a deficiency of TRIM67 exacerbated metabolic disorders in high-fat-diet-induced obese mice. We found exacerbated neuroinflammation and apoptosis in the hypothalamus of obese TRIM67 KO mice. We also found reduced BDNF in the hypothalamus, which affected the fat sympathetic nervous system innervation and contributed to lipid accumulation in adipose tissue under high-fat-diet exposure. In this study, we reveal potential implications between TRIM67 and the hypothalamic function responding to energy overuptake as well as a consideration for the therapeutic diagnosis of obesity.
Collapse
|
35
|
Hypothalamic CREB Regulates the Expression of Pomc-Processing Enzyme Pcsk2. Cells 2022; 11:cells11131996. [PMID: 35805082 PMCID: PMC9265861 DOI: 10.3390/cells11131996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/09/2022] [Accepted: 06/15/2022] [Indexed: 12/10/2022] Open
Abstract
Background: The hypothalamic proopiomelanocortin (Pomc) neurons act as first-order sensors of systemic energy stores, providing signals that regulate caloric intake and energy expenditure. In experimental obesity, dietary saturated fatty acids affect Pomc endopeptidases (PCs), resulting in the abnormal production of the neurotransmitters α-melanocyte-stimulating hormone (α-MSH) and β-endorphin, thus impacting energy balance. The cAMP response element-binding protein (CREB) is one of the transcription factors that control the expression of Pomc endopeptidases; however, it was previously unknown if dietary fats could affect CREB and consequently the expression of Pomc endopeptidases. Methods: Here, we used single-cell RNA sequencing analysis, PCR, immunoblot, ELISA and immunofluorescence histological assays to determine the impact of a high-fat diet (HFD) on the expression and function of hypothalamic CREB and its impact on the melanocortinergic system. Results: The results indicate that CREB is expressed in arcuate nucleus Pomc neurons and is activated as early as nine hours after the introduction of a high-fat diet. The inhibition of hypothalamic CREB using a short-hairpin RNA lentiviral vector resulted in increased diet-induced body-mass gain and reduced energy expenditure. This was accompanied by reduced expression of the Pomc endopeptidases, protein convertase 2, which are encoded by Pcsk2, and by the loss of the high-fat-diet-induced effect to inhibit the production of α-MSH. Conclusions: This study provides the first evidence for the involvement of CREB in the abnormal regulation of the hypothalamic Pomc endopeptidase system in experimental obesity.
Collapse
|
36
|
A Short-Term Sucrose Diet Impacts Cell Proliferation of Neural Precursors in the Adult Hypothalamus. Nutrients 2022; 14:nu14132564. [PMID: 35807744 PMCID: PMC9268421 DOI: 10.3390/nu14132564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 11/16/2022] Open
Abstract
Radial glia-like cells in the hypothalamus and dorsal vagal complex are neural precursors (NPs) located near subventricular organs: median eminence and area postrema, respectively. Their strategic position can detect blood-borne nutrients, hormones, and mitogenic signals. Hypothalamic NPs increase their proliferation with a mechanism that involves hemichannel (HC) activity. NPs can originate new neurons in response to a short-term high-fat diet as a compensatory mechanism. The effects of high carbohydrate Western diets on adult neurogenesis are unknown. Although sugars are usually consumed as sucrose, more free fructose is now incorporated into food items. Here, we studied the proliferation of both types of NPs in Sprague Dawley rats exposed to a short-term high sucrose diet (HSD) and a control diet. In tanycyte cultures, we evaluated the effects of glucose and fructose and a mix of both hexoses on HC activity. In rats fed an HSD, we observed an increase in the proliferative state of both precursors. Glucose, either in the presence or absence of fructose, but not fructose alone, induced in vitro HC activity. These results should broaden the understanding of the nutrient monitoring capacity of NPs in reacting to changes in feeding behavior, specifically to high sugar western diets.
Collapse
|
37
|
Fabianová K, Babeľová J, Fabian D, Popovičová A, Martončíková M, Raček A, Račeková E. Maternal High-Energy Diet during Pregnancy and Lactation Impairs Neurogenesis and Alters the Behavior of Adult Offspring in a Phenotype-Dependent Manner. Int J Mol Sci 2022; 23:ijms23105564. [PMID: 35628378 PMCID: PMC9146615 DOI: 10.3390/ijms23105564] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/09/2022] [Accepted: 05/13/2022] [Indexed: 11/30/2022] Open
Abstract
Obesity is one of the biggest and most costly health challenges the modern world encounters. Substantial evidence suggests that the risk of metabolic syndrome or obesity formation may be affected at a very early stage of development, in particular through fetal and/or neonatal overfeeding. Outcomes from epidemiological studies indicate that maternal nutrition during pregnancy and lactation has a profound impact on adult neurogenesis in the offspring. In the present study, an intergenerational dietary model employing overfeeding of experimental mice during prenatal and early postnatal development was applied to acquire mice with various body conditions. We investigated the impact of the maternal high-energy diet during pregnancy and lactation on adult neurogenesis in the olfactory neurogenic region involving the subventricular zone (SVZ) and the rostral migratory stream (RMS) and some behavioral tasks including memory, anxiety and nociception. Our findings show that a maternal high-energy diet administered during pregnancy and lactation modifies proliferation and differentiation, and induced degeneration of cells in the SVZ/RMS of offspring, but only in mice where extreme phenotype, such as significant overweight/adiposity or obesity is manifested. Thereafter, a maternal high-energy diet enhances anxiety-related behavior in offspring regardless of its body condition and impairs learning and memory in offspring with an extreme phenotype.
Collapse
Affiliation(s)
- Kamila Fabianová
- Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Šoltésovej 4, 040 01 Košice, Slovakia; (A.P.); (M.M.); (A.R.); (E.R.)
- Correspondence:
| | - Janka Babeľová
- Centre of Biosciences, Institute of Animal Physiology, Slovak Academy of Sciences, Šoltésovej 4-6, 040 01 Košice, Slovakia; (J.B.); (D.F.)
| | - Dušan Fabian
- Centre of Biosciences, Institute of Animal Physiology, Slovak Academy of Sciences, Šoltésovej 4-6, 040 01 Košice, Slovakia; (J.B.); (D.F.)
| | - Alexandra Popovičová
- Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Šoltésovej 4, 040 01 Košice, Slovakia; (A.P.); (M.M.); (A.R.); (E.R.)
| | - Marcela Martončíková
- Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Šoltésovej 4, 040 01 Košice, Slovakia; (A.P.); (M.M.); (A.R.); (E.R.)
| | - Adam Raček
- Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Šoltésovej 4, 040 01 Košice, Slovakia; (A.P.); (M.M.); (A.R.); (E.R.)
| | - Enikő Račeková
- Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Šoltésovej 4, 040 01 Košice, Slovakia; (A.P.); (M.M.); (A.R.); (E.R.)
| |
Collapse
|
38
|
Petermann AB, Reyna-Jeldes M, Ortega L, Coddou C, Yévenes GE. Roles of the Unsaturated Fatty Acid Docosahexaenoic Acid in the Central Nervous System: Molecular and Cellular Insights. Int J Mol Sci 2022; 23:5390. [PMID: 35628201 PMCID: PMC9141004 DOI: 10.3390/ijms23105390] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/01/2022] [Accepted: 05/04/2022] [Indexed: 11/16/2022] Open
Abstract
Fatty acids (FAs) are essential components of the central nervous system (CNS), where they exert multiple roles in health and disease. Among the FAs, docosahexaenoic acid (DHA) has been widely recognized as a key molecule for neuronal function and cell signaling. Despite its relevance, the molecular pathways underlying the beneficial effects of DHA on the cells of the CNS are still unclear. Here, we summarize and discuss the molecular mechanisms underlying the actions of DHA in neural cells with a special focus on processes of survival, morphological development, and synaptic maturation. In addition, we examine the evidence supporting a potential therapeutic role of DHA against CNS tumor diseases and tumorigenesis. The current results suggest that DHA exerts its actions on neural cells mainly through the modulation of signaling cascades involving the activation of diverse types of receptors. In addition, we found evidence connecting brain DHA and ω-3 PUFA levels with CNS diseases, such as depression, autism spectrum disorders, obesity, and neurodegenerative diseases. In the context of cancer, the existing data have shown that DHA exerts positive actions as a coadjuvant in antitumoral therapy. Although many questions in the field remain only partially resolved, we hope that future research may soon define specific pathways and receptor systems involved in the beneficial effects of DHA in cells of the CNS, opening new avenues for innovative therapeutic strategies for CNS diseases.
Collapse
Affiliation(s)
- Ana B. Petermann
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4070386, Chile;
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago 8330025, Chile; (M.R.-J.); (L.O.)
| | - Mauricio Reyna-Jeldes
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago 8330025, Chile; (M.R.-J.); (L.O.)
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica Del Norte, Coquimbo 1781421, Chile
- Núcleo para el Estudio del Cáncer a Nivel Básico, Aplicado y Clínico, Universidad Católica del Norte, Antofagasta 1270709, Chile
| | - Lorena Ortega
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago 8330025, Chile; (M.R.-J.); (L.O.)
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica Del Norte, Coquimbo 1781421, Chile
- Núcleo para el Estudio del Cáncer a Nivel Básico, Aplicado y Clínico, Universidad Católica del Norte, Antofagasta 1270709, Chile
| | - Claudio Coddou
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago 8330025, Chile; (M.R.-J.); (L.O.)
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica Del Norte, Coquimbo 1781421, Chile
- Núcleo para el Estudio del Cáncer a Nivel Básico, Aplicado y Clínico, Universidad Católica del Norte, Antofagasta 1270709, Chile
| | - Gonzalo E. Yévenes
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4070386, Chile;
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago 8330025, Chile; (M.R.-J.); (L.O.)
| |
Collapse
|
39
|
Robbins JP, Solito E. Does Neuroinflammation Underlie the Cognitive Changes Observed With Dietary Interventions? Front Neurosci 2022; 16:854050. [PMID: 35620671 PMCID: PMC9127342 DOI: 10.3389/fnins.2022.854050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
Dietary interventions, such as calorie restriction and ketogenic diet, have been extensively studied in ageing research, including in cognitive decline. Epidemiological studies indicate beneficial effects of certain dietary regimes on mental health, including mood disorders and dementia. However, randomised-controlled trials (the gold-standard of evidence-based medicine) on calorie restriction diets and the ketogenic diet have yet to show clinically convincing effects in neuropsychiatric disorders. This review will examine the quality of studies and evidence base for the ketogenic and calorie restriction diets in common neuropsychiatric conditions, collating findings from preclinical experiments, case reports or small clinical studies, and randomised controlled clinical trials. The major cellular mechanisms that mediate the effects of these dietary interventions on brain health include neuroinflammation, neuroprotection, and neuromodulation. We will discuss the studies that have investigated the roles of these pathways and their interactions. Popularity of the ketogenic and calorie restriction diets has grown both in the public domain and in psychiatry research, allowing for informed review of the efficacy, the limitations, and the side effects of these diets in specific patient populations. In this review we will summarise the clinical evidence for these diets in neuropsychiatry and make suggestions to improve clinical translation of future research studies.
Collapse
Affiliation(s)
- Jacqueline P. Robbins
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Egle Solito
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
40
|
Grasso P. Harnessing the Power of Leptin: The Biochemical Link Connecting Obesity, Diabetes, and Cognitive Decline. Front Aging Neurosci 2022; 14:861350. [PMID: 35527735 PMCID: PMC9072663 DOI: 10.3389/fnagi.2022.861350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/21/2022] [Indexed: 12/02/2022] Open
Abstract
In this review, the current understanding of leptin’s role in energy balance, glycemic regulation, and cognitive function is examined, and its involvement in maintaining the homeostatic “harmony” of these physiologies is explored. The effects of exercise on circulating leptin levels are summarized, and the results of clinical application of leptin to metabolic disease and neurologic dysfunction are reviewed. Finally, pre-clinical evidence is presented which suggests that synthetic peptide leptin mimetics may be useful in resolving not only the leptin resistance associated with common obesity and other elements of metabolic syndrome, but also the peripheral insulin resistance characterizing type 2 diabetes mellitus, and the central insulin resistance associated with certain neurologic deficits in humans.
Collapse
Affiliation(s)
- Patricia Grasso
- Department of Medicine, Albany Medical College, Albany, NY, United States
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
- *Correspondence: Patricia Grasso,
| |
Collapse
|
41
|
Fan X, Zhong Y, Zhang L, Li J, Xie F, Zhang Z. Abdominal Obesity: An Independent Influencing Factor of Visuospatial and Executive/Language Ability and the Serum Levels of A β40/A β42/Tau Protein. DISEASE MARKERS 2022; 2022:3622149. [PMID: 35401883 PMCID: PMC8993554 DOI: 10.1155/2022/3622149] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/17/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND Although obesity affects human health and cognitive function, the influence of abdominal obesity on cognitive function is still unclear. METHODS The MoCA scale was used to evaluate the overall cognitive function and the function of each subitem of 196 subjects, as well as the SDMT and TMT-A scales for evaluating the attention and information processing speed. In addition, radioimmunoassay was used to detect the serum levels of Aβ40, Aβ42, and tau protein in 45 subjects. Subjects were divided into abdominal and nonabdominal obesity groups. Before and after correcting confounding factors, the differences in cognitive scale evaluation indexes and three protein levels between the two groups were compared. We also explore further the correlation between various cognitive abilities and the waist circumference/levels of the three proteins. Linear regression was used to identify the independent influencing factors of various cognitive functions and three protein levels. RESULTS After correcting for multiple factors, we observed the lower scores of visuospatial function, execution, and language in the MoCA scale, as well as higher levels of Aβ40 and tau protein in the abdominal obesity group, supported by the results of correlation analysis. Abdominal obesity was identified as an independent negative influencing factor of MoCA visual space, executive power, and language scores and an independent positive influencing factor of Aβ40, Aβ42, and tau protein levels. CONCLUSION Abdominal obesity may play a negative role in visuospatial, executive ability, and language function and a positive role in the Aβ40, Aβ42, and tau protein serum levels.
Collapse
Affiliation(s)
- Xin Fan
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Yun Zhong
- The First Clinical Medical College of Nanchang University, Nanchang 330000, China
| | - Lingling Zhang
- School of Stomatology, Nanchang University, Nanchang 330000, China
| | - Jiaqi Li
- School of Stomatology, Nanchang University, Nanchang 330000, China
| | - Fei Xie
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Zhiyuan Zhang
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330000, China
| |
Collapse
|
42
|
Fearby N, Penman S, Thanos P. Effects of Δ9-Tetrahydrocannibinol (THC) on Obesity at Different Stages of Life: A Literature Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19063174. [PMID: 35328862 PMCID: PMC8951828 DOI: 10.3390/ijerph19063174] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/04/2022] [Indexed: 12/20/2022]
Abstract
The Cannabis sativa plant has historically been used for both recreational and medical purposes. With the recent surge in recreational use of cannabis among adolescents and adults in particular, there is an increased obligation to determine the short- and long-term effects that consuming this plant may have on several aspects of the human psyche and body. The goal of this article was to examine the negative effects of obesity, and how the use of Δ9-tetrahydrocannibinol (THC) or cannabidiol (CBD) can impact rates of this global pandemic at different timepoints of life. Conflicting studies have been reported between adult and adolescents, as there are reports of THC use leading to increased weight due to elevated appetite and consumption of food, while others observed a decrease in overall body weight due to the regulation of omega-6/omega-3 endocannabinoid precursors and a decrease in energy expenditure. Studies supported a positive correlation between prenatal cannabis use and obesity rates in the children as they matured. The data did not indicate a direct connection between prenatal THC levels in cannabis and obesity rates, but that this development may occur due to prenatal THC consumption leading to low birthweight, and subsequent obesity. There are few studies using animal models that directly measure the effects that prenatal THC administration on obesity risks among offspring. Thus, this is a critical area for future studies using a developmental framework to examine potential changes in risk across development.
Collapse
Affiliation(s)
- Nathan Fearby
- Department of Biological Sciences, University at Buffalo, Buffalo, NY 14203, USA;
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Department of Pharmacology and Toxicology, Clinical Research Institute on Addictions, Jacobs School of Medicine and Biosciences, University at Buffalo, Buffalo, NY 14203, USA;
| | - Samantha Penman
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Department of Pharmacology and Toxicology, Clinical Research Institute on Addictions, Jacobs School of Medicine and Biosciences, University at Buffalo, Buffalo, NY 14203, USA;
| | - Panayotis Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Department of Pharmacology and Toxicology, Clinical Research Institute on Addictions, Jacobs School of Medicine and Biosciences, University at Buffalo, Buffalo, NY 14203, USA;
- Department of Psychology, University at Buffalo, Buffalo, NY 14203, USA
- Correspondence: ; Tel.: +1-(716)-881-7520
| |
Collapse
|
43
|
Sa M, Park MG, Lee CJ. Role of Hypothalamic Reactive Astrocytes in Diet-Induced Obesity. Mol Cells 2022; 45:65-75. [PMID: 35236781 PMCID: PMC8907000 DOI: 10.14348/molcells.2022.2044] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 11/30/2022] Open
Abstract
Hypothalamus is a brain region that controls food intake and energy expenditure while sensing signals that convey information about energy status. Within the hypothalamus, molecularly and functionally distinct neurons work in concert under physiological conditions. However, under pathological conditions such as in diet-induced obesity (DIO) model, these neurons show dysfunctional firing patterns and distorted regulation by neurotransmitters and neurohormones. Concurrently, resident glial cells including astrocytes dramatically transform into reactive states. In particular, it has been reported that reactive astrogliosis is observed in the hypothalamus, along with various neuroinflammatory signals. However, how the reactive astrocytes control and modulate DIO by influencing neighboring neurons is not well understood. Recently, new lines of evidence have emerged indicating that these reactive astrocytes directly contribute to the pathology of obesity by synthesizing and tonically releasing the major inhibitory transmitter GABA. The released GABA strongly inhibits the neighboring neurons that control energy expenditure. These surprising findings shed light on the interplay between reactive astrocytes and neighboring neurons in the hypothalamus. This review summarizes recent discoveries related to the functions of hypothalamic reactive astrocytes in obesity and raises new potential therapeutic targets against obesity.
Collapse
Affiliation(s)
- Moonsun Sa
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
| | - Mingu Gordon Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
| | - C. Justin Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
| |
Collapse
|
44
|
Vesga-Jiménez DJ, Martin C, Barreto GE, Aristizábal-Pachón AF, Pinzón A, González J. Fatty Acids: An Insight into the Pathogenesis of Neurodegenerative Diseases and Therapeutic Potential. Int J Mol Sci 2022; 23:2577. [PMID: 35269720 PMCID: PMC8910658 DOI: 10.3390/ijms23052577] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/12/2022] [Accepted: 01/20/2022] [Indexed: 12/13/2022] Open
Abstract
One of the most common lipids in the human body is palmitic acid (PA), a saturated fatty acid with essential functions in brain cells. PA is used by cells as an energy source, besides being a precursor of signaling molecules and protein tilting across the membrane. Although PA plays physiological functions in the brain, its excessive accumulation leads to detrimental effects on brain cells, causing lipotoxicity. This mechanism involves the activation of toll-like receptors (TLR) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathways, with the consequent release of pro-inflammatory cytokines, increased production of reactive oxygen species (ROS), endoplasmic reticulum (ER) stress, and autophagy impairment. Importantly, some of the cellular changes induced by PA lead to an augmented susceptibility to the development of Alzheimer's and Parkinson´s diseases. Considering the complexity of the response to PA and the intrinsic differences of the brain, in this review, we provide an overview of the molecular and cellular effects of PA on different brain cells and their possible relationships with neurodegenerative diseases (NDs). Furthermore, we propose the use of other fatty acids, such as oleic acid or linoleic acid, as potential therapeutic approaches against NDs, as these fatty acids can counteract PA's negative effects on cells.
Collapse
Affiliation(s)
- Diego Julián Vesga-Jiménez
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogota 110231, Colombia; (D.J.V.-J.); (A.F.A.-P.)
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA 30329, USA;
| | - Cynthia Martin
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA 30329, USA;
| | - George E. Barreto
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland;
- Health Research Institute, University of Limerick, V94 T9PX Limerick, Ireland
| | - Andrés Felipe Aristizábal-Pachón
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogota 110231, Colombia; (D.J.V.-J.); (A.F.A.-P.)
| | - Andrés Pinzón
- Laboratorio de Bioinformática y Biología de Sistemas, Universidad Nacional de Colombia, Bogota 111321, Colombia;
| | - Janneth González
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogota 110231, Colombia; (D.J.V.-J.); (A.F.A.-P.)
| |
Collapse
|
45
|
Engel DF, Velloso LA. The timeline of neuronal and glial alterations in experimental obesity. Neuropharmacology 2022; 208:108983. [PMID: 35143850 DOI: 10.1016/j.neuropharm.2022.108983] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 01/03/2022] [Accepted: 02/02/2022] [Indexed: 12/14/2022]
Abstract
In experimental models, hypothalamic dysfunction is a key component of the pathophysiology of diet-induced obesity. Early after the introduction of a high-fat diet, neurons, microglia, astrocytes and tanycytes of the mediobasal hypothalamus undergo structural and functional changes that impact caloric intake, energy expenditure and systemic glucose tolerance. Inflammation has emerged as a central component of this response, and as in other inflammatory conditions, there is a time course of events that determine the fate of distinct cells involved in the central regulation of whole-body energy homeostasis. Here, we review the work that identified key mechanisms, cellular players and temporal features of diet-induced hypothalamic abnormalities.
Collapse
Affiliation(s)
- Daiane F Engel
- School of Pharmacy, Federal University of Ouro Preto, Brazil
| | - Licio A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Brazil.
| |
Collapse
|
46
|
de León-Guerrero SD, Salazar-León J, Meza-Sosa KF, Valle-Garcia D, Aguilar-León D, Pedraza-Alva G, Pérez-Martínez L. An enriched environment reestablishes metabolic homeostasis by reducing obesity-induced inflammation. Dis Model Mech 2022; 15:274225. [PMID: 35112705 PMCID: PMC9227715 DOI: 10.1242/dmm.048936] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 12/09/2021] [Indexed: 11/20/2022] Open
Abstract
Obesity can lead to chronic inflammation in different tissues, generating insulin and leptin resistance and alterations in glucose and lipid metabolism, favoring the development of degenerative diseases, including type II diabetes. Congruently, the inflammatory signaling inhibition prevents the development of obesity and restores insulin sensitivity. Via the enhancement of central nervous system activity, an enriched environment (EE) has beneficial effects on learning and memory as well as on immune cell functions and inflammation in different disease models. Here, we explored whether an EE can restore energy balance in obese mice that previously presented metabolic alterations. We discovered that an EE improved glucose metabolism, increased insulin signaling in liver, and reduced hepatic steatosis and inflammation, and increased lipolysis and browning in the white adipose tissue of high-fat diet (HFD)-fed mice. Finally, we found reduced inflammatory signaling and increased anorexigenic signaling in the hypothalamus of HFD-fed mice exposed to an EE. These data indicate that an EE is able to restore the metabolic imbalance caused by HFD feeding. Thus, we propose EE as a novel therapeutic approach for treating obesity-related metabolic alterations. This article has an associated First Person interview with the first author of the paper. Summary: A series of physiological, histochemical and molecular analyses reveal that enriched environment decreases inflammation in adipose tissue and in hypothalamus, re-establishing glucose metabolism in metabolically compromised mice.
Collapse
Affiliation(s)
- Sol Díaz de León-Guerrero
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, CP 62210, México
| | - Jonathan Salazar-León
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, CP 62210, México
| | - Karla F Meza-Sosa
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, CP 62210, México
| | - David Valle-Garcia
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, CP 62210, México
| | - Diana Aguilar-León
- Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Tlalpan, Ciudad de México, CP 14000, México
| | - Gustavo Pedraza-Alva
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, CP 62210, México
| | - Leonor Pérez-Martínez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, CP 62210, México
| |
Collapse
|
47
|
Rosenbaum JL, Melhorn SJ, Schoen S, Webb MF, De Leon MRB, Humphreys M, Utzschneider KM, Schur EA. Evidence That Hypothalamic Gliosis Is Related to Impaired Glucose Homeostasis in Adults With Obesity. Diabetes Care 2022; 45:416-424. [PMID: 34848489 PMCID: PMC8914420 DOI: 10.2337/dc21-1535] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/03/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Preclinical research implicates hypothalamic glial cell responses in the pathogenesis of obesity and type 2 diabetes (T2D). In the current study we sought to translate such findings to humans by testing whether radiologic markers of gliosis in the mediobasal hypothalamus (MBH) were greater in individuals with obesity and impaired glucose homeostasis or T2D. RESEARCH DESIGN AND METHODS Using cross-sectional and prospective cohort study designs, we applied a validated quantitative MRI approach to assess gliosis in 67 adults with obesity and normal glucose tolerance, impaired glucose tolerance (IGT), or T2D. Assessments of glucose homeostasis were conducted via oral glucose tolerance tests (OGTT) and β-cell modeling. RESULTS We found significantly greater T2 relaxation times (a marker of gliosis by MRI), that were independent of adiposity, in the groups with IGT and T2D as compared with the group with normal glucose tolerance. Findings were present in the MBH, but not control regions. Moreover, positive linear associations were present in the MBH but not control regions between T2 relaxation time and glucose area under the curve during an OGTT, fasting glucose concentrations, hemoglobin A1c, and visceral adipose tissue mass, whereas negative linear relationships were present in the MBH for markers of insulin sensitivity and β-cell function. In a prospective cohort study, greater MBH T2 relaxation times predicted declining insulin sensitivity over 1 year. CONCLUSIONS Findings support a role for hypothalamic gliosis in the progression of insulin resistance in obesity and thus T2D pathogenesis in humans.
Collapse
Affiliation(s)
- Jennifer L Rosenbaum
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA
| | - Susan J Melhorn
- Division of General Internal Medicine, Department of Medicine, University of Washington, Seattle, WA.,UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Stefan Schoen
- University of Washington School of Medicine, Seattle, WA
| | - Mary F Webb
- Division of General Internal Medicine, Department of Medicine, University of Washington, Seattle, WA.,UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Mary Rosalynn B De Leon
- Division of General Internal Medicine, Department of Medicine, University of Washington, Seattle, WA.,UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Madelaine Humphreys
- Division of General Internal Medicine, Department of Medicine, University of Washington, Seattle, WA.,UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Kristina M Utzschneider
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA.,Research and Development, Department of Veterans Affairs, Seattle, WA
| | - Ellen A Schur
- Division of General Internal Medicine, Department of Medicine, University of Washington, Seattle, WA.,UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| |
Collapse
|
48
|
Wu L, Li H, Xu W, Dong B, Geng H, Jin J, Han D, Liu H, Zhu X, Yang Y, Xie S. Emodin alleviates acute hypoxia-induced apoptosis in gibel carp (Carassius gibelio) by upregulating autophagy through modulation of the AMPK/mTOR pathway. AQUACULTURE 2022; 548:737689. [DOI: 10.1016/j.aquaculture.2021.737689] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
49
|
Macêdo APA, Gonçalves MDS, Barreto Medeiros JM, David JM, Villarreal CF, Macambira SG, Soares MBP, Couto RD. Potential therapeutic effects of green tea on obese lipid profile - a systematic review. Nutr Health 2022; 28:401-415. [PMID: 35014893 DOI: 10.1177/02601060211073236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Green tea, obtained from the plant Camellis sinensis, is one of the oldest drinks in the world and contains numerous bioactive compounds. Studies have demonstrated the efficacy of green tea in preventing obesity and cardiovascular diseases that may be related to the reduction of lipid levels. Aim: This study aimed to evidence, through a systematic review, the therapeutic potential of green tea on the lipid profile in preclinical studies in obese animals and clinical studies in obese individuals. Methods: This systematic review follows the recommendations of the preferred report items for systematic reviews and meta-analyses. The electronic databases, PubMed (Medline), Science Direct, Scopus, and Web of Science were consulted. Articles from January 2009 to December 2019 were selected. Results: This search resulted in twenty-nine articles were included cirtically reviewed. In experimental studies, green tea administration has been shown to reduce total cholesterol, triglycerides and low-density lipoprotein cholesterol in animals exposed to obesity-inducing diet. In humans' studies green tea was not shown to be effective for obese lipid control. Because supplementation with green tea extract reduced total cholesterol, triglycerides, low-density lipoprotein for three months at a specific dose. Conclusion: Therefore, green tea appears to act as a protective agent for dyslipidemia in obesity-induced animals. In human studies, green tea has not been shown to be effective in controlling obese lipids.
Collapse
Affiliation(s)
- Ana Paula Azevêdo Macêdo
- Postgraduate Program in Food Sciences, Faculty of Pharmacy, 28111Federal University of Bahia, Salvador, Bahia, Brazil
| | - Mariane Dos Santos Gonçalves
- Postgraduate Program in Food Sciences, Faculty of Pharmacy, 28111Federal University of Bahia, Salvador, Bahia, Brazil
| | | | - Jorge Mauricio David
- Department of Organic Chemistry, Institute of Chemistry, Federal University of Bahia, Salvador, Bahia, Brazil
| | | | - Simone Garcia Macambira
- Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Milena Botelho Pereira Soares
- Laboratory of Tissue Engineering and Immuno Pharmacology, 42509Research Center Gonçalo Moniz, Oswaldo Cruz Foundation, Salvador, Bahia, Brazil
| | - Ricardo David Couto
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil
| |
Collapse
|
50
|
Baraskar K, Thakur P, Shrivastava R, Shrivastava VK. Female obesity: Association with endocrine disruption and reproductive dysfunction. OBESITY MEDICINE 2021; 28:100375. [DOI: 10.1016/j.obmed.2021.100375] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|