1
|
Tirado-García P, Ferreiro A, González-Alday R, Arias-Ramos N, Lizarbe B, López-Larrubia P. Aquaporin-4 inhibition alters cerebral glucose dynamics predominantly in obese animals: an MRI study. Sci Rep 2025; 15:15649. [PMID: 40325102 PMCID: PMC12052982 DOI: 10.1038/s41598-025-99641-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025] Open
Abstract
Glucose uptake and metabolism are linked to microvascular blood flow and cellular swelling events, which are altered during obesity and can be quantified using magnetic resonance imaging (MRI). Aquaporin-4 (AQP4), the most abundant water-transporting transmembrane protein in the central nervous system, facilitates glucose transport and metabolism-derived water influx. However, its significance and regulatory capacity remain largely unknown. To better understand these processes, we acquired sequential diffusion tensor and T2*-weighted images of the brains of obese and non-obese mice, both before administering an AQP4 inhibitor and after a subsequent glucose challenge. We then subjected the resulting variables to principal component and linear mixed model analyses to assess the influence of diet, sex, administration of the inhibitor, and brain region on the data. Our findings indicate that AQP4-inhibited mice exhibit MRI values consistent with reduced microvascular blood flow and region-specific inhibition of glucose-induced cell swelling during obesity, highlighting a key role for AQP4 in glucose uptake and metabolism. Additionally, we observed that, prior to any experimental manipulation, obese mice displayed MRI signs of lower hippocampal blood flow and cerebral cellular anisotropy compared to controls, in agreement with vascular alterations and reactive gliosis processes.
Collapse
Affiliation(s)
- Pablo Tirado-García
- Institute for Biomedical Research Sols-Morreale (IIBM), Spanish National Research Council-Universidad Autónoma de Madrid, c/ Arturo Duperier 4, 28029, Madrid, Spain
| | - Adriana Ferreiro
- Institute for Biomedical Research Sols-Morreale (IIBM), Spanish National Research Council-Universidad Autónoma de Madrid, c/ Arturo Duperier 4, 28029, Madrid, Spain
| | - Raquel González-Alday
- Institute for Biomedical Research Sols-Morreale (IIBM), Spanish National Research Council-Universidad Autónoma de Madrid, c/ Arturo Duperier 4, 28029, Madrid, Spain
| | - Nuria Arias-Ramos
- Institute for Biomedical Research Sols-Morreale (IIBM), Spanish National Research Council-Universidad Autónoma de Madrid, c/ Arturo Duperier 4, 28029, Madrid, Spain
| | - Blanca Lizarbe
- Institute for Biomedical Research Sols-Morreale (IIBM), Spanish National Research Council-Universidad Autónoma de Madrid, c/ Arturo Duperier 4, 28029, Madrid, Spain.
- Department of Biochemistry, Universidad Autónoma de Madrid, Madrid, Spain.
| | - Pilar López-Larrubia
- Institute for Biomedical Research Sols-Morreale (IIBM), Spanish National Research Council-Universidad Autónoma de Madrid, c/ Arturo Duperier 4, 28029, Madrid, Spain.
- Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
2
|
Salberg S, Macowan M, Doshen A, Yamakawa GR, Sgro M, Marsland B, Henderson LA, Mychasiuk R. A high fat, high sugar diet exacerbates persistent post-surgical pain and modifies the brain-microbiota-gut axis in adolescent rats. Neuroimage 2025; 307:121057. [PMID: 39870258 DOI: 10.1016/j.neuroimage.2025.121057] [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: 08/12/2024] [Revised: 01/11/2025] [Accepted: 01/24/2025] [Indexed: 01/29/2025] Open
Abstract
Persistent post-surgical pain (PPSP) occurs in a proportion of patients following surgical interventions. Research suggests that specific microbiome components are important for brain development and function, with recent studies demonstrating that chronic pain results in changes to the microbiome. Consumption of a high fat, high sugar (HFHS) diet can drastically alter composition of the microbiome and is a modifiable risk factor for many neuroinflammatory conditions. Therefore, we investigated how daily consumption of a HFHS diet modified the development of PPSP, brain structure and function, and the microbiome. In addition, we identified significant correlations between the microbiome and brain in animals with PPSP. Male and female rats were maintained on a control or HFHS diet. Animals were further allocated to a sham or surgery on postnatal day (p) p35. The von Frey task measured mechanical nociceptive sensitivity at a chronic timepoint (p65-67). Between p68-72 rats underwent in-vivo MRI to examine brain volume and diffusivity. At p73 fecal samples were used for downstream 16 s rRNA sequencing. Spearman correlation analyses were performed between individual microbial abundance and MRI diffusivity to determine if specific bacterial species were associated with PPSP-induced brain changes. We found that consumption of a HFHS diet exacerbated PPSP in adolescents. The HFHS diet reduced overall brain volume and increased white and grey matter density. The HFHS diet interacted with the surgical intervention to modify diffusivity in numerous brain regions which were associated with specific changes to the microbiome. These findings demonstrate that premorbid characteristics can influence the development of PPSP and advance our understanding of the contribution that the microbiome has on function of the brain-microbiota-gut axis.
Collapse
Affiliation(s)
- Sabrina Salberg
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Gastroenterology, Immunology, Neuroscience (GIN) Discovery Program, Australia
| | - Matthew Macowan
- Gastroenterology, Immunology, Neuroscience (GIN) Discovery Program, Australia; Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - Angela Doshen
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, NSW, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Gastroenterology, Immunology, Neuroscience (GIN) Discovery Program, Australia
| | - Marissa Sgro
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Gastroenterology, Immunology, Neuroscience (GIN) Discovery Program, Australia
| | - Benjamin Marsland
- Gastroenterology, Immunology, Neuroscience (GIN) Discovery Program, Australia; Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - Luke A Henderson
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, NSW, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Gastroenterology, Immunology, Neuroscience (GIN) Discovery Program, Australia.
| |
Collapse
|
3
|
Huang A, Yeum D, Sewaybricker LE, Aleksic S, Thomas M, Melhorn SJ, Earley YF, Schur EA. Update on Hypothalamic Inflammation and Gliosis: Expanding Evidence of Relevance Beyond Obesity. Curr Obes Rep 2025; 14:6. [PMID: 39775194 PMCID: PMC11963668 DOI: 10.1007/s13679-024-00595-8] [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] [Accepted: 11/14/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE OF REVIEW To evaluate the role of hypothalamic inflammation and gliosis in human obesity pathogenesis and other disease processes influenced by obesity. RECENT FINDINGS Recent studies using established and novel magnetic resonance imaging (MRI) techniques to assess alterations in hypothalamic microarchitecture in humans support the presence of hypothalamic inflammation and gliosis in adults and children with obesity. Studies also identify prenatal exposure to maternal obesity or diabetes as a risk factor for hypothalamic inflammation and gliosis and increased obesity risk in offspring. Hypothalamic inflammation and gliosis have been further implicated in reproductive dysfunction (specifically polycystic ovarian syndrome and male hypogonadism), cardiovascular disease namely hypertension, and alterations in the gut microbiome, and may also accelerate neurocognitive aging. The most recent translational studies support the link between hypothalamic inflammation and gliosis and obesity pathogenesis in humans and expand our understanding of its influence on broader aspects of human health.
Collapse
Affiliation(s)
- Alyssa Huang
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Dabin Yeum
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Sandra Aleksic
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Melbin Thomas
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Susan J Melhorn
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Yumei Feng Earley
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Ellen A Schur
- Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
4
|
Correa-da-Silva F, Yi CX. Neuroglia in eating disorders (obesity, Prader-Willi syndrome and anorexia nervosa). HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:313-324. [PMID: 40148052 DOI: 10.1016/b978-0-443-19102-2.00019-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
The hypothalamus is widely recognized as one of the most extensively studied brain regions involved in the central regulation of energy homeostasis. Within the hypothalamus, peptidergic neurons play a crucial role in monitoring peripheral concentrations of metabolites and hormones, and they finely adjust the sensing of these factors, leading to the activation of either anorexigenic (appetite-suppressing) or orexigenic (appetite-stimulating) pathways. While cortical innervation of the hypothalamus does influence these processes, it is generally considered of secondary importance. Eating-related disorders, such as obesity and anorexia nervosa, are strongly associated with imbalances in energy intake and expenditure. The phenotypes of these disorders can be attributed to dysfunctions in the hypothalamus. Traditionally, it has been believed that hypothalamic dysfunction in these disorders primarily stems from defects in neural pathways. However, recent evidence challenges this perception, highlighting the active participation of neuroglial cells in shaping both physiologic and behavioral characteristics. This review aims to provide an overview of the latest insights into glial biology in three specific eating disorders: obesity, Prader-Willi syndrome, and anorexia. In these disorders, neural dysfunction coincides with glial malfunction, suggesting that neuroglia actively contribute to the development and progression of various neurologic disorders. These findings underscore the importance of glial cells and open up potential new avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Felipe Correa-da-Silva
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, The Netherlands; Laboratory of Endocrinology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, The Netherlands; Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, The Netherlands; Laboratory of Endocrinology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, The Netherlands; Netherlands Institute for Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
5
|
Solch-Ottaiano RJ, Engler-Chiurazzi EB, Harper C, Wasson S, Ogbonna S, Ouvrier B, Wang H, Prats M, McDonald K, Biose IJ, Rowe LA, Jones M, Steele C, Bix G, Maraganore DM. Comparison Between Two Divergent Diets, Mediterranean and Western, on Gut Microbiota and Cognitive Function in Young Sprague Dawley Rats. GUT MICROBES REPORTS 2024; 1:1-21. [PMID: 39916748 PMCID: PMC11800364 DOI: 10.1080/29933935.2024.2439490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 11/19/2024] [Accepted: 12/02/2024] [Indexed: 02/09/2025]
Abstract
Clinical studies strongly suggest the importance of diet quality on cognition in youth populations (15-24 years). The Mediterranean diet (MeDi) has been shown to improve cognition in contrast to the commonly consumed Western diet (WD). The gut microbiota may serve as a mechanism for diet-induced changes in cognition. Ten-week-old male Sprague Dawley rats were assigned a MeDi or WD (n=10/group) for 14 weeks. Prior to neurobehavior assessments, microbiota community composition was assessed. At the genus level, the relative abundance of four bacteria increased with the MeDi and five decreased compared to the WD. Rats in the MeDi group demonstrated cognitive flexibility and improvement in reference and working memory relative to the WD group. At the end of the study, serum cytokines were increased, and low-density lipoproteins were decreased in the MeDi group. Markers for neuroinflammation, blood-brain barrier, glial cells, and synaptic plasticity in brain regions did not differ between groups. Overall, the MeDi modulated gut microbiota, cognitive function, and serum lipid and cytokines but not gene expression in the brain compared to the WD. Further studies are needed to determine causality between diet-modulated gut microbiota, cognitive function, and immune function.
Collapse
Affiliation(s)
- Rebecca J. Solch-Ottaiano
- Clinical Neuroscience Research Center, Department of Neurology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Elizabeth B. Engler-Chiurazzi
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Colin Harper
- Clinical Neuroscience Research Center, Department of Neurology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
| | - Savannah Wasson
- Clinical Neuroscience Research Center, Department of Neurology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Sharon Ogbonna
- Clinical Neuroscience Research Center, Department of Neurology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Blake Ouvrier
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Hanyun Wang
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Madison Prats
- Clinical Neuroscience Research Center, Department of Neurology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
| | - Katherine McDonald
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ifechukwude J. Biose
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Lori A. Rowe
- Virus Characterization, Isolation, Production and Sequencing Core, Department of Microbiology, Tulane National Primate Center, Covington, LA, USA
| | - MaryJane Jones
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Chad Steele
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Gregory Bix
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Demetrius M. Maraganore
- Clinical Neuroscience Research Center, Department of Neurology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
| |
Collapse
|
6
|
Khan R, Laumet G, Leinninger GM. Hungry for relief: Potential for neurotensin to address comorbid obesity and pain. Appetite 2024; 200:107540. [PMID: 38852785 DOI: 10.1016/j.appet.2024.107540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024]
Abstract
Chronic pain and obesity frequently occur together. An ideal therapy would alleviate pain without weight gain, and most optimally, could promote weight loss. The neuropeptide neurotensin (Nts) has been separately implicated in reducing weight and pain but could it be a common actionable target for both pain and obesity? Here we review the current knowledge of Nts signaling via its receptors in modulating body weight and pain processing. Evaluating the mechanism by which Nts impacts ingestive behavior, body weight, and analgesia has potential to identify common physiologic mechanisms underlying weight and pain comorbidities, and whether Nts may be common actionable targets for both.
Collapse
Affiliation(s)
- Rabail Khan
- Neuroscience Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Geoffroy Laumet
- Neuroscience Program, Michigan State University, East Lansing, MI, 48824, USA; Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA
| | - Gina M Leinninger
- Neuroscience Program, Michigan State University, East Lansing, MI, 48824, USA; Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
7
|
Pané A, Videla L, Calvet À, Viaplana J, Vaqué-Alcázar L, Ibarzabal A, Rozalem-Aranha M, Pegueroles J, Moize V, Vidal J, Ortega E, Barroeta I, Camacho V, Chiva-Blanch G, Fortea J, Jiménez A. Hypothalamic Inflammation Improves Through Bariatric Surgery, and Hypothalamic Volume Predicts Short-Term Weight Loss Response in Adults With or Without Type 2 Diabetes. Diabetes Care 2024; 47:1162-1170. [PMID: 38713908 DOI: 10.2337/dc23-2213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 04/03/2024] [Indexed: 05/09/2024]
Abstract
OBJECTIVE Preclinical research implicates hypothalamic inflammation (HI) in obesity and type 2 diabetes pathophysiology. However, their pathophysiological relevance and potential reversibility need to be better defined. We sought to evaluate the effect of bariatric surgery (BS) on radiological biomarkers of HI and the association between the severity of such radiological alterations and post-BS weight loss (WL) trajectories. The utility of cerebrospinal fluid large extracellular vesicles (CSF-lEVs) enriched for microglial and astrocyte markers in studying HI was also explored. RESEARCH DESIGN AND METHODS We included 72 individuals with obesity (20 with and 52 without type 2 diabetes) and 24 control individuals. Participants underwent lumbar puncture and 3-T MRI at baseline and 1-year post-BS. We assessed hypothalamic mean diffusivity (MD) (higher values indicate lesser microstructural integrity) and the volume of the whole and main hypothalamic subregions. CSF-lEVs enriched for glial and astrocyte markers were determined by flow cytometry. RESULTS Compared with control group, the obesity and type 2 diabetes groups showed a larger volume and higher MD in the hypothalamic tubular inferior region, the area encompassing the arcuate nucleus. These radiological alterations were positively associated with baseline anthropometric and metabolic measures and improved post-BS. A larger baseline tubular inferior hypothalamic volume was independently related to lesser WL 1 and 2 years after BS. CSF-lEVs did not differ among groups and were unrelated to WL trajectories. CONCLUSIONS These findings suggest HI improvement after BS and may support a role for HI in modulating the WL response to these interventions.
Collapse
Affiliation(s)
- Adriana Pané
- Endocrinology and Nutrition Department, Hospital Clínic, Barcelona, Spain
- CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Videla
- Sant Pau Memory Unit, Neurology Department, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- CIBERNED, Instituto de Salud Carlos III
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Àngels Calvet
- Fundació Clínic per la Recerca Biomèdica-Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain
| | - Judith Viaplana
- Fundació Clínic per la Recerca Biomèdica-Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain
| | - Lídia Vaqué-Alcázar
- Sant Pau Memory Unit, Neurology Department, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Fundació Clínic per la Recerca Biomèdica-Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain
- Department of Medicine, Faculty of Medicine and Health Sciences and Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Ainitze Ibarzabal
- Gastrointestinal Surgery Department, Hospital Clínic, Barcelona, Spain
| | - Mateus Rozalem-Aranha
- Sant Pau Memory Unit, Neurology Department, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jordi Pegueroles
- Sant Pau Memory Unit, Neurology Department, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Violeta Moize
- Endocrinology and Nutrition Department, Hospital Clínic, Barcelona, Spain
- Fundació Clínic per la Recerca Biomèdica-Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain
| | - Josep Vidal
- Endocrinology and Nutrition Department, Hospital Clínic, Barcelona, Spain
- Fundació Clínic per la Recerca Biomèdica-Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain
- CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
| | - Emilio Ortega
- Endocrinology and Nutrition Department, Hospital Clínic, Barcelona, Spain
- CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
- Fundació Clínic per la Recerca Biomèdica-Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain
| | - Isabel Barroeta
- Sant Pau Memory Unit, Neurology Department, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- CIBERNED, Instituto de Salud Carlos III
| | - Valle Camacho
- Nuclear Medicine Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Gemma Chiva-Blanch
- CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
- Fundació Clínic per la Recerca Biomèdica-Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain
- Health Sciences Faculty, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Neurology Department, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- CIBERNED, Instituto de Salud Carlos III
| | - Amanda Jiménez
- Endocrinology and Nutrition Department, Hospital Clínic, Barcelona, Spain
- CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
- Fundació Clínic per la Recerca Biomèdica-Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain
| |
Collapse
|
8
|
Frago LM, Gómez-Romero A, Collado-Pérez R, Argente J, Chowen JA. Synergism Between Hypothalamic Astrocytes and Neurons in Metabolic Control. Physiology (Bethesda) 2024; 39:0. [PMID: 38530221 DOI: 10.1152/physiol.00009.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/05/2024] [Accepted: 03/22/2024] [Indexed: 03/27/2024] Open
Abstract
Astrocytes are no longer considered as passive support cells. In the hypothalamus, these glial cells actively participate in the control of appetite, energy expenditure, and the processes leading to obesity and its secondary complications. Here we briefly review studies supporting this conclusion and the advances made in understanding the underlying mechanisms.
Collapse
Affiliation(s)
- Laura M Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Alfonso Gómez-Romero
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Roberto Collado-Pérez
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, Campus of International Excellence, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, Campus of International Excellence, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
9
|
Salvi J, Andreoletti P, Audinat E, Balland E, Ben Fradj S, Cherkaoui-Malki M, Heurtaux T, Liénard F, Nédélec E, Rovère C, Savary S, Véjux A, Trompier D, Benani A. Microgliosis: a double-edged sword in the control of food intake. FEBS J 2024; 291:615-631. [PMID: 35880408 DOI: 10.1111/febs.16583] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/30/2022] [Accepted: 07/25/2022] [Indexed: 02/16/2024]
Abstract
Maintaining energy balance is essential for survival and health. This physiological function is controlled by the brain, which adapts food intake to energy needs. Indeed, the brain constantly receives a multitude of biological signals that are derived from digested foods or that originate from the gastrointestinal tract, energy stores (liver and adipose tissues) and other metabolically active organs (muscles). These signals, which include circulating nutrients, hormones and neuronal inputs from the periphery, collectively provide information on the overall energy status of the body. In the brain, several neuronal populations can specifically detect these signals. Nutrient-sensing neurons are found in discrete brain areas and are highly enriched in the hypothalamus. In turn, specialized brain circuits coordinate homeostatic responses acting mainly on appetite, peripheral metabolism, activity and arousal. Accumulating evidence shows that hypothalamic microglial cells located at the vicinity of these circuits can influence the brain control of energy balance. However, microglial cells could have opposite effects on energy balance, that is homeostatic or detrimental, and the conditions for this shift are not totally understood yet. One hypothesis relies on the extent of microglial activation, and nutritional lipids can considerably change it.
Collapse
Affiliation(s)
- Juliette Salvi
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Pierre Andreoletti
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Etienne Audinat
- IGF, Université de Montpellier, CNRS, Inserm, Montpellier, France
| | - Eglantine Balland
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Notting Hill, Australia
| | - Selma Ben Fradj
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Valbonne, France
| | | | - Tony Heurtaux
- Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg
- Department of Life Sciences and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Fabienne Liénard
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Emmanuelle Nédélec
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Carole Rovère
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Valbonne, France
| | - Stéphane Savary
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Anne Véjux
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Doriane Trompier
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Alexandre Benani
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
10
|
Kim JD, Copperi F, Diano S. Microglia in Central Control of Metabolism. Physiology (Bethesda) 2024; 39:0. [PMID: 37962895 PMCID: PMC11283896 DOI: 10.1152/physiol.00021.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/12/2023] [Accepted: 11/12/2023] [Indexed: 11/15/2023] Open
Abstract
Beyond their role as brain immune cells, microglia act as metabolic sensors in response to changes in nutrient availability, thus playing a role in energy homeostasis. This review highlights the evidence and challenges of studying the role of microglia in metabolism regulation.
Collapse
Affiliation(s)
- Jung Dae Kim
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, New York, United States
| | - Francesca Copperi
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, New York, United States
| | - Sabrina Diano
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, New York, United States
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, New York, United States
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, New York, United States
| |
Collapse
|
11
|
Puliani G, Sbardella E, Cozzolino A, Sada V, Tozzi R, Andreoli C, Fiorelli M, Di Biasi C, Corallino D, Balla A, Paganini AM, Venneri MA, Lenzi A, Lubrano C, Isidori AM. Pituitary T1 signal intensity at magnetic resonance imaging is reduced in patients with obesity: results from the CHIASM study. Int J Obes (Lond) 2023; 47:948-955. [PMID: 37479795 PMCID: PMC10511316 DOI: 10.1038/s41366-023-01338-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 06/18/2023] [Accepted: 07/04/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND Despite obesity being well known to be associated with several pituitary hormone imbalances, pituitary appearance in magnetic resonance imaging (MRI) in patients with obesity is understudied. OBJECTIVE To evaluate the pituitary volume and signal intensity at MRI in patients with obesity. METHODS This is a prospective study performed in an endocrine Italian referral center (ClinicalTrial.gov Identifier: NCT03458533). Sixty-nine patients with obesity (BMI > 30 kg/m2) and twenty-five subjects without obesity were enrolled. Thirty-three patients with obesity were re-evaluated after 3 years of diet and lifestyle changes, of whom 17 (51.5%) achieved a > 5% loss of their initial body weight, whereas the remaining 16 (48.5%) had maintained or gained weight. Evaluations included metabolic and hormone assessments, DEXA scan, and pituitary MRI. Pituitary signal intensity was quantified by measuring the pixel density using ImageJ software. RESULTS At baseline, no difference in pituitary volume was observed between the obese and non-obese cohorts. At the 3-year follow-up, pituitary volume was significantly reduced (p = 0.011) only in participants with stable-increased body weight. Furthermore, a significant difference was noted in the mean pituitary intensity of T1-weighted plain and contrast-enhanced sequences between the obese and non-obese cohorts at baseline (p = 0.006; p = 0.002), and a significant decrease in signal intensity was observed in the subgroup of participants who had not lost weight (p = 0.012; p = 0.017). Insulin-like growth factor-1 levels, following correction for BMI, were correlated with pituitary volume (p = 0.001) and intensity (p = 0.049), whereas morning cortisol levels were correlated with pituitary intensity (p = 0.007). The T1-weighted pituitary intensity was negatively correlated with truncal fat (p = 0.006) and fibrinogen (p = 0.018). CONCLUSIONS The CHIASM study describes a quantitative reduction in pituitary intensity in T1-weighted sequences in patients with obesity. These alterations could be explained by changes in the pituitary stromal tissue, correlated with low-grade inflammation.
Collapse
Affiliation(s)
- Giulia Puliani
- Oncological Endocrinology Unit, Regina Elena National Cancer Institute IRCCS, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Emilia Sbardella
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Alessia Cozzolino
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Valentina Sada
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Rossella Tozzi
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Chiara Andreoli
- Unit of Emergency Radiology, Department of Radiological, Oncological and Pathological Sciences, Umberto I University Hospital, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - Marco Fiorelli
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università, 30, 00185, Rome, Italy
| | - Claudio Di Biasi
- Unit of Emergency Radiology, Department of Radiological, Oncological and Pathological Sciences, Umberto I University Hospital, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - Diletta Corallino
- Bariatric Surgery Unit, Department of General Surgery and Surgical Specialties "Paride Stefanini" Sapienza University of Rome, Umberto I University Hospital, Viale del Policlinico 155, 00161, Rome, Italy
| | - Andrea Balla
- Bariatric Surgery Unit, Department of General Surgery and Surgical Specialties "Paride Stefanini" Sapienza University of Rome, Umberto I University Hospital, Viale del Policlinico 155, 00161, Rome, Italy
| | - Alessandro M Paganini
- Bariatric Surgery Unit, Department of General Surgery and Surgical Specialties "Paride Stefanini" Sapienza University of Rome, Umberto I University Hospital, Viale del Policlinico 155, 00161, Rome, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Carla Lubrano
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
12
|
Douglass JD, Ness KM, Valdearcos M, Wyse-Jackson A, Dorfman MD, Frey JM, Fasnacht RD, Santiago OD, Niraula A, Banerjee J, Robblee M, Koliwad SK, Thaler JP. Obesity-associated microglial inflammatory activation paradoxically improves glucose tolerance. Cell Metab 2023; 35:1613-1629.e8. [PMID: 37572666 PMCID: PMC10528677 DOI: 10.1016/j.cmet.2023.07.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/09/2023] [Accepted: 07/19/2023] [Indexed: 08/14/2023]
Abstract
Hypothalamic gliosis associated with high-fat diet (HFD) feeding increases susceptibility to hyperphagia and weight gain. However, the body-weight-independent contribution of microglia to glucose regulation has not been determined. Here, we show that reducing microglial nuclear factor κB (NF-κB) signaling via cell-specific IKKβ deletion exacerbates HFD-induced glucose intolerance despite reducing body weight and adiposity. Conversely, two genetic approaches to increase microglial pro-inflammatory signaling (deletion of an NF-κB pathway inhibitor and chemogenetic activation through a modified Gq-coupled muscarinic receptor) improved glucose tolerance independently of diet in both lean and obese rodents. Microglial regulation of glucose homeostasis involves a tumor necrosis factor alpha (TNF-α)-dependent mechanism that increases activation of pro-opiomelanocortin (POMC) and other hypothalamic glucose-sensing neurons, ultimately leading to a marked amplification of first-phase insulin secretion via a parasympathetic pathway. Overall, these data indicate that microglia regulate glucose homeostasis in a body-weight-independent manner, an unexpected mechanism that limits the deterioration of glucose tolerance associated with obesity.
Collapse
Affiliation(s)
- John D Douglass
- UW Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Kelly M Ness
- UW Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Martin Valdearcos
- The Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alice Wyse-Jackson
- UW Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Mauricio D Dorfman
- UW Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Jeremy M Frey
- UW Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Rachael D Fasnacht
- UW Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Olivia D Santiago
- UW Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Anzela Niraula
- UW Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Jineta Banerjee
- UW Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Megan Robblee
- The Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Suneil K Koliwad
- The Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Joshua P Thaler
- UW Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
13
|
Hamamah S, Amin A, Al-Kassir AL, Chuang J, Covasa M. Dietary Fat Modulation of Gut Microbiota and Impact on Regulatory Pathways Controlling Food Intake. Nutrients 2023; 15:3365. [PMID: 37571301 PMCID: PMC10421457 DOI: 10.3390/nu15153365] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Obesity is a multifactorial disease that continues to increase in prevalence worldwide. Emerging evidence has shown that the development of obesity may be influenced by taxonomic shifts in gut microbiota in response to the consumption of dietary fats. Further, these alterations in gut microbiota have been shown to promote important changes in satiation signals including gut hormones (leptin, ghrelin, GLP-1, peptide YY and CCK) and orexigenic and anorexigenic neuropeptides (AgRP, NPY, POMC, CART) that influence hyperphagia and therefore obesity. In this review, we highlight mechanisms by which gut microbiota can influence these satiation signals both locally in the gastrointestinal tract and via microbiota-gut-brain communication. Then, we describe the effects of dietary interventions and associated changes in gut microbiota on satiety signals through microbiota-dependent mechanisms. Lastly, we present microbiota optimizing therapies including prebiotics, probiotics, synbiotics and weight loss surgery that can help restore beneficial gut microbiota by enhancing satiety signals to reduce hyperphagia and subsequent obesity. Overall, a better understanding of the mechanisms by which dietary fats induce taxonomical shifts in gut microbiota and their impact on satiation signaling pathways will help develop more targeted therapeutic interventions in delaying the onset of obesity and in furthering its treatment.
Collapse
Affiliation(s)
- Sevag Hamamah
- Department of Basic Medical Sciences, Western University of Health Sciences, College of Osteopathic Medicine, Pomona, CA 91766, USA; (S.H.); (A.A.); (A.L.A.-K.); (J.C.)
| | - Arman Amin
- Department of Basic Medical Sciences, Western University of Health Sciences, College of Osteopathic Medicine, Pomona, CA 91766, USA; (S.H.); (A.A.); (A.L.A.-K.); (J.C.)
| | - Abdul Latif Al-Kassir
- Department of Basic Medical Sciences, Western University of Health Sciences, College of Osteopathic Medicine, Pomona, CA 91766, USA; (S.H.); (A.A.); (A.L.A.-K.); (J.C.)
| | - Judith Chuang
- Department of Basic Medical Sciences, Western University of Health Sciences, College of Osteopathic Medicine, Pomona, CA 91766, USA; (S.H.); (A.A.); (A.L.A.-K.); (J.C.)
| | - Mihai Covasa
- Department of Basic Medical Sciences, Western University of Health Sciences, College of Osteopathic Medicine, Pomona, CA 91766, USA; (S.H.); (A.A.); (A.L.A.-K.); (J.C.)
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania
| |
Collapse
|
14
|
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
|
15
|
Obesity-Induced Brain Neuroinflammatory and Mitochondrial Changes. Metabolites 2023; 13:metabo13010086. [PMID: 36677011 PMCID: PMC9865135 DOI: 10.3390/metabo13010086] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 12/31/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023] Open
Abstract
Obesity is defined as abnormal and excessive fat accumulation, and it is a risk factor for developing metabolic and neurodegenerative diseases and cognitive deficits. Obesity is caused by an imbalance in energy homeostasis resulting from increased caloric intake associated with a sedentary lifestyle. However, the entire physiopathology linking obesity with neurodegeneration and cognitive decline has not yet been elucidated. During the progression of obesity, adipose tissue undergoes immune, metabolic, and functional changes that induce chronic low-grade inflammation. It has been proposed that inflammatory processes may participate in both the peripheral disorders and brain disorders associated with obesity, including the development of cognitive deficits. In addition, mitochondrial dysfunction is related to inflammation and oxidative stress, causing cellular oxidative damage. Preclinical and clinical studies of obesity and metabolic disorders have demonstrated mitochondrial brain dysfunction. Since neuronal cells have a high energy demand and mitochondria play an important role in maintaining a constant energy supply, impairments in mitochondrial activity lead to neuronal damage and dysfunction and, consequently, to neurotoxicity. In this review, we highlight the effect of obesity and high-fat diet consumption on brain neuroinflammation and mitochondrial changes as a link between metabolic dysfunction and cognitive decline.
Collapse
|
16
|
Deng Y, Yi X, Gong Y, Zhou L, Xie D, Wang J, Liu Z, Zhang Y, Wu W. Palmitic acid induces nDNA release to cytosol and promotes microglial M1 polarization via cGAS-STING signaling pathway. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119385. [PMID: 36302463 DOI: 10.1016/j.bbamcr.2022.119385] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 10/08/2022] [Accepted: 10/16/2022] [Indexed: 11/06/2022]
Abstract
Palmitic acid (PA), the most common statured fatty acid in diets, is involved in peripheral as well as central inflammation. The M1 polarization of microglia plays an important role in PA-induced neuroinflammation. However, it is still unclear on the key factor and molecule mechanism of microglial polarization among it. Thus, we investigated whether the release of self-DNA into the cytoplasm of microglia was a consequence of PA treatment, as in aortic endothelial cells and adipocytes. RT-qPCR and immunofluorescence were performed to detect the status of cytosolic DNA and microglial polarization after PA treatment. We found that the content of cytosolic nDNA rather than mtDNA increased after PA treatment and the M1 polarization of microglia was associated with this. Moreover, the knockdown of cGAS in BV2 microglial cells demonstrated that the cGAS-STING pathway is involved in polarization process. Our results revealed that nDNA and cGAS-STING pathway are critically involved in PA-induced microglial M1 polarization. This mechanism may pose a new insight on targeting microglia may be a promising way to mitigate diet-induced early neuroinflammation.
Collapse
Affiliation(s)
- Yuping Deng
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaoqing Yi
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yuxiang Gong
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Liyan Zhou
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Dongxue Xie
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jufen Wang
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhilin Liu
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yinhao Zhang
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wenhe Wu
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
17
|
Trends in Gliosis in Obesity, and the Role of Antioxidants as a Therapeutic Alternative. Antioxidants (Basel) 2022; 11:antiox11101972. [PMID: 36290695 PMCID: PMC9598641 DOI: 10.3390/antiox11101972] [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: 08/24/2022] [Revised: 09/23/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
Obesity remains a global health problem. Chronic low-grade inflammation in this pathology has been related to comorbidities such as cognitive alterations that, in the long term, can lead to neurodegenerative diseases. Neuroinflammation or gliosis in patients with obesity and type 2 diabetes mellitus has been related to the effect of adipokines, high lipid levels and glucose, which increase the production of free radicals. Cerebral gliosis can be a risk factor for developing neurodegenerative diseases, and antioxidants could be an alternative for the prevention and treatment of neural comorbidities in obese patients. AIM Identify the immunological and oxidative stress mechanisms that produce gliosis in patients with obesity and propose antioxidants as an alternative to reducing neuroinflammation. METHOD Advanced searches were performed in scientific databases: PubMed, ProQuest, EBSCO, and the Science Citation index for research on the physiopathology of gliosis in obese patients and for the possible role of antioxidants in its management. CONCLUSION Patients with obesity can develop neuroinflammation, conditioned by various adipokines, excess lipids and glucose, which results in an increase in free radicals that must be neutralized with antioxidants to reduce gliosis and the risk of long-term neurodegeneration.
Collapse
|
18
|
Steuernagel L, Lam BYH, Klemm P, Dowsett GKC, Bauder CA, Tadross JA, Hitschfeld TS, Del Rio Martin A, Chen W, de Solis AJ, Fenselau H, Davidsen P, Cimino I, Kohnke SN, Rimmington D, Coll AP, Beyer A, Yeo GSH, Brüning JC. HypoMap-a unified single-cell gene expression atlas of the murine hypothalamus. Nat Metab 2022; 4:1402-1419. [PMID: 36266547 PMCID: PMC9584816 DOI: 10.1038/s42255-022-00657-y] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 09/07/2022] [Indexed: 01/20/2023]
Abstract
The hypothalamus plays a key role in coordinating fundamental body functions. Despite recent progress in single-cell technologies, a unified catalog and molecular characterization of the heterogeneous cell types and, specifically, neuronal subtypes in this brain region are still lacking. Here, we present an integrated reference atlas, 'HypoMap,' of the murine hypothalamus, consisting of 384,925 cells, with the ability to incorporate new additional experiments. We validate HypoMap by comparing data collected from Smart-Seq+Fluidigm C1 and bulk RNA sequencing of selected neuronal cell types with different degrees of cellular heterogeneity. Finally, via HypoMap, we identify classes of neurons expressing glucagon-like peptide-1 receptor (Glp1r) and prepronociceptin (Pnoc), and validate them using single-molecule in situ hybridization. Collectively, HypoMap provides a unified framework for the systematic functional annotation of murine hypothalamic cell types, and it can serve as an important platform to unravel the functional organization of hypothalamic neurocircuits and to identify druggable targets for treating metabolic disorders.
Collapse
Affiliation(s)
- Lukas Steuernagel
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Brian Y H Lam
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Paul Klemm
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Georgina K C Dowsett
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Corinna A Bauder
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - John A Tadross
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
- Cambridge Genomics Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Tamara Sotelo Hitschfeld
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Almudena Del Rio Martin
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Weiyi Chen
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Alain J de Solis
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Henning Fenselau
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany
| | | | - Irene Cimino
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Sara N Kohnke
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Debra Rimmington
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Anthony P Coll
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Andreas Beyer
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
- Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany.
| | - Giles S H Yeo
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK.
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany.
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany.
- National Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
19
|
Lama A, Pirozzi C, Severi I, Morgese MG, Senzacqua M, Annunziata C, Comella F, Del Piano F, Schiavone S, Petrosino S, Mollica MP, Diano S, Trabace L, Calignano A, Giordano A, Mattace Raso G, Meli R. Palmitoylethanolamide dampens neuroinflammation and anxiety-like behavior in obese mice. Brain Behav Immun 2022; 102:110-123. [PMID: 35176443 PMCID: PMC10662208 DOI: 10.1016/j.bbi.2022.02.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 02/01/2022] [Accepted: 02/04/2022] [Indexed: 12/11/2022] Open
Abstract
High-fat diet (HFD) consumption leads to obesity and a chronic state of low-grade inflammation, named metainflammation. Notably, metainflammation contributes to neuroinflammation due to the increased levels of circulating free fatty acids and cytokines. It indicates a strict interplay between peripheral and central counterparts in the pathogenic mechanisms of obesity-related mood disorders. In this context, the impairment of internal hypothalamic circuitry runs in tandem with the alteration of other brain areas associated with emotional processing (i.e., hippocampus and amygdala). Palmitoylethanolamide (PEA), an endogenous lipid mediator belonging to the N-acylethanolamines family, has been extensively studied for its pleiotropic effects both at central and peripheral level. Our study aimed to elucidate PEA capability in limiting obesity-induced anxiety-like behavior and neuroinflammation-related features in an experimental model of HFD-fed obese mice. PEA treatment promoted an improvement in anxiety-like behavior of obese mice and the systemic inflammation, reducing serum pro-inflammatory mediators (i.e., TNF-α, IL-1β, MCP-1, LPS). In the amygdala, PEA increased dopamine turnover, as well as GABA levels. PEA also counteracted the overactivation of HPA axis, reducing the expression of hypothalamic corticotropin-releasing hormone and its type 1 receptor. Moreover, PEA attenuated the immunoreactivity of Iba-1 and GFAP and reduced pro-inflammatory pathways and cytokine production in both the hypothalamus and hippocampus. This finding, together with the reduced transcription of mast cell markers (chymase 1 and tryptase β2) in the hippocampus, indicated the weakening of immune cell activation underlying the neuroprotective effect of PEA. Obesity-driven neuroinflammation was also associated with the disruption of blood-brain barrier (BBB) in the hippocampus. PEA limited the albumin extravasation and restored tight junction transcription modified by HFD. To gain mechanistic insight, we designed an in vitro model of metabolic injury using human neuroblastoma SH-SY5Y cells insulted by a mix of glucosamine and glucose. Here, PEA directly counteracted inflammation and mitochondrial dysfunction in a PPAR-α-dependent manner since the pharmacological blockade of the receptor reverted its effects. Our results strengthen the therapeutic potential of PEA in obesity-related neuropsychiatric comorbidities, controlling neuroinflammation, BBB disruption, and neurotransmitter imbalance involved in behavioral dysfunctions.
Collapse
Affiliation(s)
- Adriano Lama
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 49 - 80131 Naples, Italy
| | - Claudio Pirozzi
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 49 - 80131 Naples, Italy
| | - Ilenia Severi
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto, 10, A - 60020 Ancona, Italy
| | - Maria Grazia Morgese
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20 - 71122 Foggia, Italy
| | - Martina Senzacqua
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto, 10, A - 60020 Ancona, Italy
| | - Chiara Annunziata
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 49 - 80131 Naples, Italy
| | - Federica Comella
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 49 - 80131 Naples, Italy
| | - Filomena Del Piano
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Via Delpino 1, 80137 Naples, Italy
| | - Stefania Schiavone
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20 - 71122 Foggia, Italy
| | - Stefania Petrosino
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Napoli, Italy
| | - Maria Pina Mollica
- Department of Biology, University of Naples Federico II, Complesso Universitario di Monte, Sant'Angelo, Cupa Nuova Cinthia 21 - Edificio 7, 80126 Naples, Italy
| | - Sabrina Diano
- Program in Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20 - 71122 Foggia, Italy
| | - Antonio Calignano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 49 - 80131 Naples, Italy
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto, 10, A - 60020 Ancona, Italy
| | - Giuseppina Mattace Raso
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 49 - 80131 Naples, Italy.
| | - Rosaria Meli
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 49 - 80131 Naples, Italy
| |
Collapse
|
20
|
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
|
21
|
Effect of Propionic Acid on Diabetes-Induced Impairment of Unfolded Protein Response Signaling and Astrocyte/Microglia Crosstalk in Rat Ventromedial Nucleus of the Hypothalamus. Neural Plast 2022; 2022:6404964. [PMID: 35103058 PMCID: PMC8800605 DOI: 10.1155/2022/6404964] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/17/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
Abstract
Background The aim was to investigate the influence of propionic acid (PA) on the endoplasmic reticulum (ER), unfolded protein response (UPR) state, and astrocyte/microglia markers in rat ventromedial hypothalamus (VMH) after type 2 diabetes mellitus (T2DM). Methods Male Wistar rats were divided: (1) control, (2) T2DM, and groups that received the following (14 days, orally): (3) metformin (60 mg/kg), (4) PA (60 mg/kg), and (5) PA+metformin. Western blotting, RT-PCR, transmission electron microscopy, and immunohistochemical staining were performed. Results We found T2DM-associated enlargement of ER cisterns, while drug administration slightly improved VMH ultrastructural signs of damage. GRP78 level was 2.1-fold lower in T2DM vs. control. Metformin restored GRP78 to control, while PA increased it by 2.56-fold and metformin+PA—by 3.28-fold vs. T2DM. PERK was elevated by 3.61-fold in T2DM, after metformin—by 4.98-fold, PA—5.64-fold, and metformin+PA—3.01-fold vs. control. A 2.45-fold increase in ATF6 was observed in T2DM. Metformin decreased ATF6 content vs. T2DM. Interestingly, PA exerted a more pronounced lowering effect on ATF6, while combined treatment restored ATF6 to control. IRE1 increased in T2DM (2.4-fold), metformin (1.99-fold), and PA (1.45-fold) groups vs. control, while metformin+PA fully normalized its content. The Iba1 level was upregulated in T2DM (5.44-fold) and metformin groups (6.88-fold). Despite PA treatment leading to a further 8.9-fold Iba1 elevation, PA+metformin caused the Iba1 decline vs. metformin and PA treatment. GFAP level did not change in T2DM but rose in metformin and PA groups vs. control. PA+metformin administration diminished GFAP vs. PA. T2DM-induced changes were associated with dramatically decreased ZO-1 levels, while PA treatment increased it almost to control values. Conclusions T2DM-induced UPR imbalance, activation of microglia, and impairments in cell integrity may trigger VMH dysfunction. Drug administration slightly improved ultrastructural changes in VMH, normalized UPR, and caused an astrocyte activation. PA and metformin exerted beneficial effects for counteracting diabetes-induced ER stress in VMH.
Collapse
|
22
|
Bhusal A, Rahman MH, Suk K. Hypothalamic inflammation in metabolic disorders and aging. Cell Mol Life Sci 2021; 79:32. [PMID: 34910246 PMCID: PMC11071926 DOI: 10.1007/s00018-021-04019-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/01/2021] [Accepted: 10/29/2021] [Indexed: 12/15/2022]
Abstract
The hypothalamus is a critical brain region for the regulation of energy homeostasis. Over the years, studies on energy metabolism primarily focused on the neuronal component of the hypothalamus. Studies have recently uncovered the vital role of glial cells as an additional player in energy balance regulation. However, their inflammatory activation under metabolic stress condition contributes to various metabolic diseases. The recruitment of monocytes and macrophages in the hypothalamus helps sustain such inflammation and worsens the disease state. Neurons were found to actively participate in hypothalamic inflammatory response by transmitting signals to the surrounding non-neuronal cells. This activation of different cell types in the hypothalamus leads to chronic, low-grade inflammation, impairing energy balance and contributing to defective feeding habits, thermogenesis, and insulin and leptin signaling, eventually leading to metabolic disorders (i.e., diabetes, obesity, and hypertension). The hypothalamus is also responsible for the causation of systemic aging under metabolic stress. A better understanding of the multiple factors contributing to hypothalamic inflammation, the role of the different hypothalamic cells, and their crosstalks may help identify new therapeutic targets. In this review, we focus on the role of glial cells in establishing a cause-effect relationship between hypothalamic inflammation and the development of metabolic diseases. We also cover the role of other cell types and discuss the possibilities and challenges of targeting hypothalamic inflammation as a valid therapeutic approach.
Collapse
Affiliation(s)
- Anup Bhusal
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Md Habibur Rahman
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
- Division of Endocrinology, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea.
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea.
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41944, Republic of Korea.
| |
Collapse
|
23
|
Neves TMG, Simoes E, Otaduy MCG, Calfat ELDB, Bertolazzi P, da Costa NA, Duran FLDS, Correia-Lima J, Martin MDGM, Seelander MCL, Otani VHO, Otani TZDS, Vasques DAC, Filho GB, Kochi C, Uchida RR. Inverse Association Between Hypothalamic N-Acetyl Aspartate/Creatine Ratio and Indices of Body Mass in Adolescents with Obesity. J Nutr 2021; 152:663-670. [PMID: 34888674 PMCID: PMC8891176 DOI: 10.1093/jn/nxab415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/06/2021] [Accepted: 12/03/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Approximately 10% of adolescents worldwide are overweight or obese, hence the urgent and universal need to elucidate possible mechanisms that lead to obesity in the adolescent population. OBJECTIVES We examined the hypothalamic metabolism and its relationship with physical development in obese and eutrophic adolescents. METHODS We performed a case-control study with 115 adolescents between 11 and 18 years of age, to compare obese (BMI z-score ≥ 2) and nonobese individuals (eutrophic controls; BMI z-score ≤ 1). The following hypothalamic metabolite ratios were examined as primary outcomes: glutamate/creatine (Cr), the sum of glutamate and glutamine/Cr, N-acetylaspartate (NAA)/Cr, myoinositol/Cr, and total choline/Cr (glycerophosphocholine + phosphocholine/Cr), quantified by magnetic resonance spectroscopy. BMI z-scores, pubertal status, and scores on the Yale Food Addiction Scale, the Binge Eating Scale, and the Child Depression Inventory were assessed as secondary outcomes. Pearson coefficients (r) or nonparametric Spearman correlation (rho) analyses were performed between hypothalamic metabolite ratios and other parameters, such as BMI z-scores, physical development, food habits, depression symptoms, and serum protein concentrations (cytokines, hormones, and neuropeptides). RESULTS Adolescents with obesity showed a lower hypothalamic NAA/Cr ratio (0.70 ± 0.19) compared to their eutrophic counterparts (0.84 ± 0.20; P = 0.004). The NAA/Cr ratio was negatively correlated with BMI z-scores (r = -0.25; P = 0.03) and serum insulin (rho = -0.27; P = 0.04), C-peptide (rho = -0.26; P = 0.04), amylin (r = -0.27; P = 0.04), ghrelin (rho = -0.30; P = 0.02), and neuropeptide Y (r = -0.27; P = 0.04). Also, the NAA/Cr ratio was positively correlated with circulating IL-8 levels (rho = 0.26; P = 0.04). CONCLUSIONS High BMI z-scores are associated with lower hypothalamic NAA/Cr ratios. The negative correlations found between the NAA/Cr ratio and serum cytokines, hormones, and neuropeptides suggest a broad cross-talk linking hormonal imbalances, neurohumoral alterations, and hypothalamic functions in adolescents with obesity.
Collapse
Affiliation(s)
| | | | | | | | - Pâmela Bertolazzi
- Mental Health Department, Santa Casa de Sao Paulo School of Medical Sciences, São Paulo, Brazil
| | - Naomi Antunes da Costa
- Neuroimaging Laboratory (LIM-21), Institute Psychiatry, University of São Paulo, São Paulo, Brazil
| | | | - Joanna Correia-Lima
- Cancer Metabolism Research Group, University of São Paulo, São Paulo, Brazil
| | | | - Marília Cerqueira Leite Seelander
- Cancer Metabolism Research Group, University of São Paulo, São Paulo, Brazil,Department of Surgery and LIM 26, Hospital das Clínicas, University of São Paulo, São Paulo, Brazil
| | | | | | | | - Geraldo Busatto Filho
- Neuroimaging Laboratory (LIM-21), Institute Psychiatry, University of São Paulo, São Paulo, Brazil
| | - Cristiane Kochi
- Pediatrics Department, Santa Casa de Sao Paulo School of Medical Sciences, São Paulo, Brazil
| | - Ricardo Riyoiti Uchida
- Mental Health Department, Santa Casa de Sao Paulo School of Medical Sciences, São Paulo, Brazil
| |
Collapse
|
24
|
Sewaybricker LE, Kee S, Melhorn SJ, Schur EA. Greater radiologic evidence of hypothalamic gliosis predicts adiposity gain in children at risk for obesity. Obesity (Silver Spring) 2021; 29:1770-1779. [PMID: 34734493 PMCID: PMC8608399 DOI: 10.1002/oby.23286] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/22/2021] [Accepted: 08/10/2021] [Indexed: 12/26/2022]
Abstract
OBJECTIVE This study investigated, in a large pediatric population, whether magnetic resonance imaging (MRI) evidence of mediobasal hypothalamic (MBH) gliosis is associated with baseline or change over 1 year in body adiposity. METHODS Cross-sectional and prospective cohort analyses were conducted within the Adolescent Brain Cognitive Development Study. Study 1 included 169 children with usable baseline T2-weighted MRI images and anthropometrics from baseline and 1-year follow-up study visits. Signal ratios compared T2 signal intensity in MBH and two reference regions (amygdala [AMY] and putamen) as a measure of MBH gliosis. Study 2 included a distinct group of 238 children with overweight or obesity to confirm initial findings in an independent sample. RESULTS In Study 1, MBH/AMY signal ratio was positively associated with BMI z score (β = 4.27, p < 0.001). A significant interaction for the association of MBH/AMY signal ratio with change in BMI z score suggested that relationships differed by baseline weight status. Study 2 found that higher MBH/AMY signal ratios associated with an increase in BMI z score for children with overweight (β = 0.58, p = 0.01), but not those with obesity (β = 0.02, p = 0.91). CONCLUSIONS Greater evidence of hypothalamic gliosis by MRI is associated with baseline BMI z score and predicts adiposity gain in young children at risk of obesity.
Collapse
Affiliation(s)
| | - Sarah Kee
- Dept. of Medicine, University of Washington, Seattle, WA, USA
| | | | - Ellen A. Schur
- Dept. of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
25
|
Long-term diet-induced obesity does not lead to learning and memory impairment in adult mice. PLoS One 2021; 16:e0257921. [PMID: 34587222 PMCID: PMC8480843 DOI: 10.1371/journal.pone.0257921] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 09/13/2021] [Indexed: 02/06/2023] Open
Abstract
Obesity arising from excessive dietary fat intake is a risk factor for cognitive decline, dementia and neurodegenerative diseases, including Alzheimer’s disease. Here, we studied the effect of long-term high-fat diet (HFD) (24 weeks) and return to normal diet (ND) on behavioral features, microglia and neurons in adult male C57BL/6J mice. Consequences of HFD-induced obesity and dietary changes on general health (coat appearance, presence of vibrissae), sensory and motor reflexes, learning and memory were assessed by applying a phenotypic assessment protocol, the Y maze and Morris Water Maze test. Neurons and microglia were histologically analyzed within the mediobasal hypothalamus, hippocampus and frontal motor cortex after long-term HFD and change of diet. Long periods of HFD caused general health issues (coat alterations, loss of vibrissae), but did not affect sensory and motor reflexes, emotional state, memory and learning. Long-term HFD increased the microglial response (increased Iba1 fluorescence intensity, percentage of Iba1-stained area and Iba1 gene expression) within the hypothalamus, but not in the cortex and hippocampus. In neither of these regions, neurodegeneration or intracellular lipid droplet accumulation was observed. The former alterations were reversible in mice whose diet was changed from HFD to ND. Taken together, long periods of excessive dietary fat alone do not cause learning deficits or spatial memory impairment, though HFD-induced obesity may have detrimental consequences for cognitive flexibility. Our data confirm the selective responsiveness of hypothalamic microglia to HFD.
Collapse
|
26
|
Ebrahim HA, El-Gamal R, Sherif RN. Intermittent Fasting Attenuates High Fat Diet-Induced Cerebellar Changes in Rats: Involvement of TNFα, autophagy and oxidative stress. Cells Tissues Organs 2021; 210:351-367. [PMID: 34551416 DOI: 10.1159/000519088] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/12/2021] [Indexed: 11/19/2022] Open
Affiliation(s)
- Hasnaa Ali Ebrahim
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
- Department of Anatomy, Faculty of Medicine, University of Mansoura, Mansoura, Egypt
| | - Randa El-Gamal
- Department of Medical Biochemistry, Faculty of Medicine, University of Mansoura, Mansoura, Egypt
- Medical Experimental Research Center (MERC), Faculty of Medicine, University of Mansoura, Mansoura, Egypt
| | - Rania N Sherif
- Department of Anatomy, Faculty of Medicine, University of Mansoura, Mansoura, Egypt
- Department of Anatomy and Embryology, Faculty of Dentistry, Horus University, Damietta, Egypt
- Department of Anatomy and Embryology, Faculty of Medicine, Damietta University, Damietta, Egypt
| |
Collapse
|
27
|
Ye L, Jia G, Li Y, Wang Y, Chen H, Yu L, Wu D. C1q/TNF-related protein 4 restores leptin sensitivity by downregulating NF-κB signaling and microglial activation. J Neuroinflammation 2021; 18:159. [PMID: 34275474 PMCID: PMC8286609 DOI: 10.1186/s12974-021-02167-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/05/2021] [Indexed: 12/29/2022] Open
Abstract
Objective C1qTNF-related protein 4 (CTRP4) acts in the hypothalamus to modulate food intake in diet-induced obese mice and has been shown to exert an anti-inflammatory effect on macrophages. Since high-fat diet-induced microglial activation and hypothalamic inflammation impair leptin signaling and increase food intake, we aimed to explore the potential connection between the anorexigenic effect of CTRP4 and the suppression of hypothalamic inflammation in mice with DIO. Methods Using an adenovirus-mediated hypothalamic CTRP4 overexpression model, we investigated the impact of CTRP4 on food intake and the hypothalamic leptin signaling pathway in diet-induced obese mice. Furthermore, central and plasma proinflammatory cytokines, including TNF-α and IL-6, were measured by Western blotting and ELISA. Changes in the hypothalamic NF-κB signaling cascade and microglial activation were also examined in vivo. In addition, NF-κB signaling and proinflammatory factors were investigated in BV-2 cells after CTRP4 intervention. Results We found that food intake was decreased, while leptin signaling was significantly improved in mice with DIO after CTRP4 overexpression. Central and peripheral TNF-α and IL-6 levels were reduced by central Ad-CTRP4 administration. Hypothalamic NF-κB signaling and microglial activation were also significantly suppressed in vivo. In addition, NF-κB signaling was inhibited in BV-2 cells following CTRP4 intervention, which was consistent with the decreased production of TNF-α and IL-6. Conclusions Our data indicate that CTRP4 reverses leptin resistance by inhibiting NF-κB-dependent microglial activation and hypothalamic inflammation. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02167-2.
Collapse
Affiliation(s)
- Liu Ye
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Gongwei Jia
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Yuejie Li
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Ying Wang
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Hong Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lehua Yu
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Dandong Wu
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
28
|
Hypothalamic Astrocytes as a Specialized and Responsive Cell Population in Obesity. Int J Mol Sci 2021; 22:ijms22126176. [PMID: 34201099 PMCID: PMC8228119 DOI: 10.3390/ijms22126176] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022] Open
Abstract
Astrocytes are a type of glial cell anatomically and functionally integrated into the neuronal regulatory circuits for the neuroendocrine control of metabolism. Being functional integral compounds of synapses, astrocytes are actively involved in the physiological regulatory aspects of metabolic control, but also in the pathological processes that link neuronal dysfunction and obesity. Between brain areas, the hypothalamus harbors specialized functional circuits that seem selectively vulnerable to metabolic damage, undergoing early cellular rearrangements which are thought to be at the core of the pathogenesis of diet-induced obesity. Such changes in the hypothalamic brain region consist of a rise in proinflammatory cytokines, the presence of a reactive phenotype in astrocytes and microglia, alterations in the cytoarchitecture and synaptology of hypothalamic circuits, and angiogenesis, a phenomenon that cannot be found elsewhere in the brain. Increasing evidence points to the direct involvement of hypothalamic astrocytes in such early metabolic disturbances, thus moving the study of these glial cells to the forefront of obesity research. Here we provide a comprehensive review of the most relevant findings of molecular and pathophysiological mechanisms by which hypothalamic astrocytes might be involved in the pathogenesis of obesity.
Collapse
|
29
|
Lyra E Silva NM, Gonçalves RA, Pascoal TA, Lima-Filho RAS, Resende EDPF, Vieira ELM, Teixeira AL, de Souza LC, Peny JA, Fortuna JTS, Furigo IC, Hashiguchi D, Miya-Coreixas VS, Clarke JR, Abisambra JF, Longo BM, Donato J, Fraser PE, Rosa-Neto P, Caramelli P, Ferreira ST, De Felice FG. Pro-inflammatory interleukin-6 signaling links cognitive impairments and peripheral metabolic alterations in Alzheimer's disease. Transl Psychiatry 2021; 11:251. [PMID: 33911072 PMCID: PMC8080782 DOI: 10.1038/s41398-021-01349-z] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/25/2021] [Accepted: 03/19/2021] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is associated with memory impairment and altered peripheral metabolism. Mounting evidence indicates that abnormal signaling in a brain-periphery metabolic axis plays a role in AD pathophysiology. The activation of pro-inflammatory pathways in the brain, including the interleukin-6 (IL-6) pathway, comprises a potential point of convergence between memory dysfunction and metabolic alterations in AD that remains to be better explored. Using T2-weighted magnetic resonance imaging (MRI), we observed signs of probable inflammation in the hypothalamus and in the hippocampus of AD patients when compared to cognitively healthy control subjects. Pathological examination of post-mortem AD hypothalamus revealed the presence of hyperphosphorylated tau and tangle-like structures, as well as parenchymal and vascular amyloid deposits surrounded by astrocytes. T2 hyperintensities on MRI positively correlated with plasma IL-6, and both correlated inversely with cognitive performance and hypothalamic/hippocampal volumes in AD patients. Increased IL-6 and suppressor of cytokine signaling 3 (SOCS3) were observed in post-mortem AD brains. Moreover, activation of the IL-6 pathway was observed in the hypothalamus and hippocampus of AD mice. Neutralization of IL-6 and inhibition of the signal transducer and activator of transcription 3 (STAT3) signaling in the brains of AD mouse models alleviated memory impairment and peripheral glucose intolerance, and normalized plasma IL-6 levels. Collectively, these results point to IL-6 as a link between cognitive impairment and peripheral metabolic alterations in AD. Targeting pro-inflammatory IL-6 signaling may be a strategy to alleviate memory impairment and metabolic alterations in the disease.
Collapse
Affiliation(s)
- Natalia M Lyra E Silva
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Rafaella A Gonçalves
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Tharick A Pascoal
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Ricardo A S Lima-Filho
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Elisa de Paula França Resende
- Behavioral and Cognitive Neurology Research Group, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Erica L M Vieira
- Centre of Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Antonio L Teixeira
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Santa Casa BH Ensino e Pesquisa, Belo Horizonte, MG, Brazil
| | - Leonardo C de Souza
- Behavioral and Cognitive Neurology Research Group, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Julyanna A Peny
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Juliana T S Fortuna
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Isadora C Furigo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Debora Hashiguchi
- Department of Physiology, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Vivian S Miya-Coreixas
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Julia R Clarke
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Jose F Abisambra
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease University of Florida, Gainesville, FL, USA
| | - Beatriz M Longo
- Department of Physiology, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Paulo Caramelli
- Behavioral and Cognitive Neurology Research Group, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada.
- Department of Psychiatry, Queen's University, Kingston, ON, Canada.
- Department of Biomedical and Molecuar Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
30
|
Loos RJF, Burant C, Schur EA. Strategies to Understand the Weight-Reduced State: Genetics and Brain Imaging. Obesity (Silver Spring) 2021; 29 Suppl 1:S39-S50. [PMID: 33759393 PMCID: PMC8500189 DOI: 10.1002/oby.23101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 11/09/2022]
Abstract
Most individuals with obesity or overweight have difficulty maintaining weight loss. The weight-reduced state induces changes in many physiological processes that appear to drive weight regain. Here, we review the use of cell biology, genetics, and imaging techniques that are being used to begin understanding why weight regain is the normal response to dieting. As with obesity itself, weight regain has both genetic and environmental drivers. Genetic drivers for "thinness" and "obesity" largely overlap, but there is evidence for specific genetic loci that are different for each of these weight states. There is only limited information regarding the genetics of weight regain. Currently, most genetic loci related to weight point to the central nervous system as the organ responsible for determining the weight set point. Neuroimaging tools have proved useful in studying the contribution of the central nervous system to the weight-reduced state in humans. Neuroimaging technologies fall into three broad categories: functional, connectivity, and structural neuroimaging. Connectivity and structural imaging techniques offer unique opportunities for testing mechanistic hypotheses about changes in brain function or tissue structure in the weight-reduced state.
Collapse
Affiliation(s)
- Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Charles Burant
- Department of Internal Medicine, University of Washington, Seattle, Washington, USA
| | - Ellen A. Schur
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
31
|
Mapping of Microglial Brain Region, Sex and Age Heterogeneity in Obesity. Int J Mol Sci 2021; 22:ijms22063141. [PMID: 33808700 PMCID: PMC8003547 DOI: 10.3390/ijms22063141] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/27/2022] Open
Abstract
The prevalence of obesity has increased rapidly in recent years and has put a huge burden on healthcare worldwide. Obesity is associated with an increased risk for many comorbidities, such as cardiovascular diseases, type 2 diabetes and hypertension. The hypothalamus is a key brain region involved in the regulation of food intake and energy expenditure. Research on experimental animals has shown neuronal loss, as well as microglial activation in the hypothalamus, due to dietary-induced obesity. Microglia, the resident immune cells in the brain, are responsible for maintaining the brain homeostasis and, thus, providing an optimal environment for neuronal function. Interestingly, in obesity, microglial cells not only get activated in the hypothalamus but in other brain regions as well. Obesity is also highly associated with changes in hippocampal function, which could ultimately result in cognitive decline and dementia. Moreover, changes have also been reported in the striatum and cortex. Microglial heterogeneity is still poorly understood, not only in the context of brain region but, also, age and sex. This review will provide an overview of the currently available data on the phenotypic differences of microglial innate immunity in obesity, dependent on brain region, sex and age.
Collapse
|
32
|
Eide PK, Hasan‐Olive MM, Hansson H, Enger R. Increased occurrence of pathological mitochondria in astrocytic perivascular endfoot processes and neurons of idiopathic intracranial hypertension. J Neurosci Res 2021; 99:467-480. [PMID: 33105056 PMCID: PMC7821105 DOI: 10.1002/jnr.24743] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
Idiopathic intracranial hypertension (IIH) primarily affects fertile, overweight women, and presents with the symptoms of raised intracranial pressure. The etiology is unknown but has been thought to relate to cerebrospinal fluid disturbance or cerebral venous stenosis. We have previously found evidence that IIH is also a disease of the brain parenchyma, evidenced by alterations at the neurogliovascular interface, including astrogliosis, pathological changes in the basement membrane and pericytes, and alterations of perivascular aquaporin-4. The aim of this present electron microscopic study was to examine whether mitochondria phenotype was changed in IIH, particularly focusing on perivascular astrocytic endfeet and neurons (soma and pre- and postsynaptic terminals). Cortical brain biopsies of nine reference individuals and eight IIH patients were analyzed for subcellular distribution and phenotypical features of mitochondria using transmission electron microscopy. We found significantly increased prevalence of pathological mitochondria and reduced number of normal mitochondria in astrocytic endfeet of IIH patients. The degree of astrogliosis correlated negatively with the number of normal mitochondria in astrocytic endfoot processes. Moreover, we found significantly increased number of pathological mitochondria in pre- and postsynaptic neuronal terminals, as well as significantly shortened distance between mitochondria and endoplasmic reticulum contacts. Finally, the length of postsynaptic density, a marker of synaptic strength, was on average reduced in IIH. The present data provide evidence of pathological mitochondria in perivascular astrocytes endfeet and neurons of IIH patients, highlighting that impaired metabolism at the neurogliovascular interface may be a facet of IIH.
Collapse
Affiliation(s)
- Per Kristian Eide
- Department of NeurosurgeryOslo University Hospital ‐ RikshospitaletOsloNorway
- Institute of Clinical MedicineFaculty of MedicineUniversity of OsloOsloNorway
| | - Md Mahdi Hasan‐Olive
- Department of NeurosurgeryOslo University Hospital ‐ RikshospitaletOsloNorway
- Institute of Clinical MedicineFaculty of MedicineUniversity of OsloOsloNorway
| | | | - Rune Enger
- GliaLab and Letten CentreDivision of Anatomy and Division of PhysiologyDepartment of Molecular MedicineInstitute of Basic Medical SciencesUniversity of OsloOsloNorway
- Department of NeurologyOslo University Hospital ‐ RikshospitaletOsloNorway
| |
Collapse
|
33
|
Robb JL, Morrissey NA, Weightman Potter PG, Smithers HE, Beall C, Ellacott KLJ. Immunometabolic Changes in Glia - A Potential Role in the Pathophysiology of Obesity and Diabetes. Neuroscience 2020; 447:167-181. [PMID: 31765625 PMCID: PMC7567742 DOI: 10.1016/j.neuroscience.2019.10.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 12/15/2022]
Abstract
Chronic low-grade inflammation is a feature of the pathophysiology of obesity and diabetes in the CNS as well as peripheral tissues. Glial cells are critical mediators of the response to inflammation in the brain. Key features of glia include their metabolic flexibility, sensitivity to changes in the CNS microenvironment, and ability to rapidly adapt their function accordingly. They are specialised cells which cooperate to promote and preserve neuronal health, playing important roles in regulating the activity of neuronal networks across the brain during different life stages. Increasing evidence points to a role of glia, most notably astrocytes and microglia, in the systemic regulation of energy and glucose homeostasis in the course of normal physiological control and during disease. Inflammation is an energetically expensive process that requires adaptive changes in cellular metabolism and, in turn, metabolic intermediates can also have immunomodulatory actions. Such "immunometabolic" changes in peripheral immune cells have been implicated in contributing to disease pathology in obesity and diabetes. This review will discuss the evidence for a role of immunometabolic changes in glial cells in the systemic regulation of energy and glucose homeostasis, and how this changes in the context of obesity and diabetes.
Collapse
Affiliation(s)
- Josephine L Robb
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Nicole A Morrissey
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Paul G Weightman Potter
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Hannah E Smithers
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Craig Beall
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Kate L J Ellacott
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK.
| |
Collapse
|
34
|
Lizarbe B, Campillo B, Guadilla I, López-Larrubia P, Cerdán S. Magnetic resonance assessment of the cerebral alterations associated with obesity development. J Cereb Blood Flow Metab 2020; 40:2135-2151. [PMID: 32703110 PMCID: PMC7585928 DOI: 10.1177/0271678x20941263] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/28/2020] [Accepted: 06/14/2020] [Indexed: 12/15/2022]
Abstract
Obesity is a current threat to health care systems, affecting approximately 13% of the world's adult population, and over 18% children and adolescents. The rise of obesity is fuelled by inadequate life style habits, as consumption of diets rich in fats and sugars which promote, additionally, the development of associated comorbidities. Obesity results from a neuroendocrine imbalance in the cerebral mechanisms controlling food intake and energy expenditure, including the hypothalamus and the reward and motivational centres. Specifically, high-fat diets are known to trigger an early inflammatory response in the hypothalamus that precedes weight gain, is time-dependent, and eventually extends to the remaining appetite regulating regions in the brain. Multiple magnetic resonance imaging (MRI) and spectroscopy (MRS) methods are currently available to characterize different features of cerebral obesity, including diffusion weighted, T2 and volumetric imaging and 1H and 13C spectroscopic evaluations. In particular, consistent evidences have revealed increased water diffusivity and T2 values, decreased grey matter volumes, and altered metabolic profiles and fluxes, in the brain of animal models and in obese humans. This review provides an integrative interpretation of the physio-pathological processes associated with obesity development in the brain, and the MRI and MRS methods implemented to characterize them.
Collapse
Affiliation(s)
- Blanca Lizarbe
- Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC/UAM, Madrid, Spain
| | - Basilio Campillo
- Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC/UAM, Madrid, Spain
| | - Irene Guadilla
- Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC/UAM, Madrid, Spain
| | | | - Sebastián Cerdán
- Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC/UAM, Madrid, Spain
| |
Collapse
|
35
|
Foright RM, Johnson GC, Kahn D, Charleston CA, Presby DM, Bouchet CA, Wellberg EA, Sherk VD, Jackman MR, Greenwood BN, MacLean PS. Compensatory eating behaviors in male and female rats in response to exercise training. Am J Physiol Regul Integr Comp Physiol 2020; 319:R171-R183. [PMID: 32551825 PMCID: PMC7473893 DOI: 10.1152/ajpregu.00259.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 04/29/2020] [Accepted: 06/09/2020] [Indexed: 12/22/2022]
Abstract
Exercise is often used as a strategy for weight loss maintenance. In preclinical models, we have shown that exercise may be beneficial because it counters the biological drive to regain weight. However, our studies have demonstrated sex differences in the response to exercise in this context. In the present study, we sought to better understand why females and males exhibit different compensatory food eating behaviors in response to regular exercise. Using a forced treadmill exercise paradigm, we measured weight gain, energy expenditure, food intake in real time, and the anorectic effects of leptin. The 4-wk exercise training resulted in reduced weight gain in males and sustained weight gain in females. In male rats, exercise decreased intake, whereas it increased food intake in females. Our results suggest that the anorectic effects of leptin were not responsible for these sex differences in appetite in response to exercise. If these results translate to the human condition, they may reveal important information for the use and application of regular exercise programs.
Collapse
Affiliation(s)
- Rebecca M Foright
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Ginger C Johnson
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Darcy Kahn
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Catherine A Charleston
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - David M Presby
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Courtney A Bouchet
- Department of Psychology, University of Colorado Denver, Denver, Colorado
| | - Elizabeth A Wellberg
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Vanessa D Sherk
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Matthew R Jackman
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | - Paul S MacLean
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
36
|
Huang HT, Chen PS, Kuo YM, Tzeng SF. Intermittent peripheral exposure to lipopolysaccharide induces exploratory behavior in mice and regulates brain glial activity in obese mice. J Neuroinflammation 2020; 17:163. [PMID: 32450884 PMCID: PMC7249324 DOI: 10.1186/s12974-020-01837-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/03/2020] [Indexed: 12/23/2022] Open
Abstract
Background Consecutive peripheral immune challenges can modulate the responses of brain resident microglia to stimuli. High-fat diet (HFD) intake has been reported to stimulate the activation of astrocytes and microglia in the arcuate nucleus (ARC) of the hypothalamus in obese rodents and humans. However, it is unknown whether intermittent exposure to additional peripheral immune challenge can modify HFD-induced hypothalamic glial activation in obese individuals. Methods In this study, we administered 1 mg/kg LPS (or saline) by intraperitoneal (i.p.) injection to 8-week-old male mice after 1, 2, or 8 weeks of a regular diet (show) or HFD. The level of interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) expression in the plasma and hypothalamic tissue was analyzed 24 h after each LPS injection. The behaviors of the animals in the four groups (the chow-saline, chow-LPS, HFD-saline, and HFD-LPS groups) were examined 5 months after exposure to chow or a HFD. Morphological examination of microglia in related brain regions was also conducted. Results The plasma levels and hypothalamic mRNA levels of IL-1β and TNF-α were significantly upregulated 24 h after the first injection of LPS but not after the second or third injection of LPS. Chow-LPS mice displayed increased exploratory behavior 5 months after feeding. However, this LPS-induced abnormal exploratory behavior was inhibited in HFD-fed mice. Chronic HFD feeding for 5 months induced apparent increases in the number and cell body size of microglia, mainly in the ARC, and also increased the size of microglia in the nucleus accumbens (NAc) and insula. Moreover, microglial activation in the ARC, anterior cingulate cortex (ACC), insula, and basolateral amygdala (BLA) was observed in chow-LPS mice. However, microglial activation in the analyzed brain regions was suppressed in HFD-LPS mice. Conclusions Altogether, the results indicate that intermittent peripheral challenge with LPS might prime microglia in the ARC and NAc to modify their response to chronic HFD feeding. Alternatively, chronic HFD feeding might mediate microglia in LPS-affected brain regions and subsequently suppress LPS-induced atypical exploratory behavior. Our findings suggest that the interaction of intermittent acute peripheral immune challenges with chronic HFD intake can drive microglia to amend the microenvironment and further modify animal behaviors in the later life.
Collapse
Affiliation(s)
- Hui-Ting Huang
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Po-See Chen
- Department of Psychiatry, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Min Kuo
- Institute of Basic Medical Sciences, Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shun-Fen Tzeng
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan. .,Department of Life Sciences, National Cheng Kung University, #1 University Road, Tainan, Taiwan.
| |
Collapse
|
37
|
Nyamugenda E, Griffin H, Russell S, Cooney KA, Kowalczyk NS, Islam I, Phelan KD, Baldini G. Selective Survival of Sim1/MC4R Neurons in Diet-Induced Obesity. iScience 2020; 23:101114. [PMID: 32438321 PMCID: PMC7240135 DOI: 10.1016/j.isci.2020.101114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/19/2020] [Accepted: 04/27/2020] [Indexed: 12/12/2022] Open
Abstract
In the melanocortin pathway, melanocortin-4 receptor (MC4R) functions to control energy homeostasis. MC4R is expressed in a sub-population of Sim1 neurons (Sim1/MC4R neurons) and functions in hypothalamic paraventricular nuclei (PVN) to control food intake. Mapping sites of hypothalamic injury in obesity is essential to counteract the disease. In the PVN of male and female mice with diet-induced obesity (DIO) there is neuronal loss. However, the existing subpopulation of PVN Sim1/MC4R neurons is unchanged, but has a loss of mitochondria and MC4R protein. In mice of both sexes with DIO, dietary intervention to re-establish normal weight restores abundance of MC4R protein in Sim1/MC4R neurons and neurogenesis in the PVN. However, the number of non-Sim1/MC4R neurons in the PVN continues to remain decreased. Selective survival and recovery of Sim1/MC4R neurons after DIO suggests these neurons as preferential target to restore energy homeostasis and of therapy against obesity.
Collapse
Affiliation(s)
- Eugene Nyamugenda
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Haven Griffin
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Susan Russell
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Kimberly A Cooney
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Nicholas S Kowalczyk
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ishrar Islam
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Kevin D Phelan
- Department of Neurobiology & 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.
| |
Collapse
|
38
|
Dorsal vagal complex and hypothalamic glia differentially respond to leptin and energy balance dysregulation. Transl Psychiatry 2020; 10:90. [PMID: 32152264 PMCID: PMC7062837 DOI: 10.1038/s41398-020-0767-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/18/2020] [Accepted: 02/25/2020] [Indexed: 01/16/2023] Open
Abstract
Previous studies identify a role for hypothalamic glia in energy balance regulation; however, a narrow hypothalamic focus provides an incomplete understanding of how glia throughout the brain respond to and regulate energy homeostasis. We examined the responses of glia in the dorsal vagal complex (DVC) to the adipokine leptin and high fat diet-induced obesity. DVC astrocytes functionally express the leptin receptor; in vivo pharmacological studies suggest that DVC astrocytes partly mediate the anorectic effects of leptin in lean but not diet-induced obese rats. Ex vivo calcium imaging indicated that these changes were related to a lower proportion of leptin-responsive cells in the DVC of obese versus lean animals. Finally, we investigated DVC microglia and astroglia responses to leptin and energy balance dysregulation in vivo: obesity decreased DVC astrogliosis, whereas the absence of leptin signaling in Zucker rats was associated with extensive astrogliosis in the DVC and decreased hypothalamic micro- and astrogliosis. These data uncover a novel functional heterogeneity of astrocytes in different brain nuclei of relevance to leptin signaling and energy balance regulation.
Collapse
|
39
|
Thomas K, Beyer F, Lewe G, Zhang R, Schindler S, Schönknecht P, Stumvoll M, Villringer A, Witte AV. Higher body mass index is linked to altered hypothalamic microstructure. Sci Rep 2019; 9:17373. [PMID: 31758009 PMCID: PMC6874651 DOI: 10.1038/s41598-019-53578-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/01/2019] [Indexed: 01/23/2023] Open
Abstract
Animal studies suggest that obesity-related diets induce structural changes in the hypothalamus, a key brain area involved in energy homeostasis. Whether this translates to humans is however largely unknown. Using a novel multimodal approach with manual segmentation, we here show that a higher body mass index (BMI) selectively predicted higher proton diffusivity within the hypothalamus, indicative of compromised microstructure in the underlying tissue, in a well-characterized population-based cohort (n1 = 338, 48% females, age 21-78 years, BMI 18-43 kg/m²). Results were independent from confounders and confirmed in another independent sample (n2 = 236). In addition, while hypothalamic volume was not associated with obesity, we identified a sexual dimorphism and larger hypothalamic volumes in the left compared to the right hemisphere. Using two large samples of the general population, we showed that a higher BMI specifically relates to altered microstructure in the hypothalamus, independent from confounders such as age, sex and obesity-associated co-morbidities. This points to persisting microstructural changes in a key regulatory area of energy homeostasis occurring with excessive weight. Our findings may help to better understand the pathomechanisms of obesity and other eating-related disorders.
Collapse
Affiliation(s)
- K Thomas
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, 04103, Leipzig, Germany
- Collaborative Research Centre 1052'Obesity Mechanisms', Subproject A1, Faculty of Medicine, Leipzig University, 04103, Leipzig, Germany
| | - F Beyer
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, 04103, Leipzig, Germany
- Collaborative Research Centre 1052'Obesity Mechanisms', Subproject A1, Faculty of Medicine, Leipzig University, 04103, Leipzig, Germany
| | - G Lewe
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, 04103, Leipzig, Germany
| | - R Zhang
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, 04103, Leipzig, Germany
| | - S Schindler
- Department of Psychiatry and Psychotherapy, Leipzig University Hospital, 04103, Leipzig, Germany
- Department of Neurophysics, Max Planck Institute for Human Cognitive and Brain Sciences, 04103, Leipzig, Germany
| | - P Schönknecht
- Department of Psychiatry and Psychotherapy, Leipzig University Hospital, 04103, Leipzig, Germany
| | - M Stumvoll
- Collaborative Research Centre 1052'Obesity Mechanisms', Subproject A1, Faculty of Medicine, Leipzig University, 04103, Leipzig, Germany
- Department of Endocrinology and Nephrology, Leipzig University Hospital, 04103, Leipzig, Germany
| | - A Villringer
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, 04103, Leipzig, Germany
- Collaborative Research Centre 1052'Obesity Mechanisms', Subproject A1, Faculty of Medicine, Leipzig University, 04103, Leipzig, Germany
- Clinic of Cognitive Neurology, Leipzig University Hospital, 04103, Leipzig, Germany
- Leipzig Research Center for Civilization Diseases (LIFE), Leipzig University, 04103, Leipzig, Germany
| | - A V Witte
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, 04103, Leipzig, Germany.
- Collaborative Research Centre 1052'Obesity Mechanisms', Subproject A1, Faculty of Medicine, Leipzig University, 04103, Leipzig, Germany.
| |
Collapse
|
40
|
Hankir MK, Rullmann M, Seyfried F, Preusser S, Poppitz S, Heba S, Gousias K, Hoyer J, Schütz T, Dietrich A, Müller K, Pleger B. Roux-en-Y gastric bypass surgery progressively alters radiologic measures of hypothalamic inflammation in obese patients. JCI Insight 2019; 4:131329. [PMID: 31465301 DOI: 10.1172/jci.insight.131329] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/23/2019] [Indexed: 12/19/2022] Open
Abstract
There is increased interest in whether bariatric surgeries such as Roux-en-Y gastric bypass (RYGB) achieve their profound weight-lowering effects in morbidly obese individuals through the brain. Hypothalamic inflammation is a well-recognized etiologic factor in obesity pathogenesis and so represents a potential target of RYGB, but clinical evidence in support of this is limited. We therefore assessed hypothalamic T2-weighted signal intensities (T2W SI) and fractional anisotropy (FA) values, 2 validated radiologic measures of brain inflammation, in relation to BMI and fat mass, as well as circulating inflammatory (C-reactive protein; CrP) and metabolic markers in a cohort of 27 RYGB patients at baseline and 6 and 12 months after surgery. We found that RYGB progressively increased hypothalamic T2W SI values, while it progressively decreased hypothalamic FA values. Regression analyses further revealed that this could be most strongly linked to plasma CrP levels, which independently predicted hypothalamic FA values when adjusting for age, sex, fat mass, and diabetes diagnosis. These findings suggest that RYGB has a major time-dependent impact on hypothalamic inflammation status, possibly by attenuating peripheral inflammation. They also suggest that hypothalamic FA values may provide a more specific radiologic measure of hypothalamic inflammation than more commonly used T2W SI values.
Collapse
Affiliation(s)
- Mohammed K Hankir
- Department of Experimental Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Michael Rullmann
- IFB AdiposityDiseases and.,Department of Nuclear Medicine, University Hospital Leipzig, Germany.,Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.,Collaborative Research Centre 1052 in Obesity Mechanisms, University of Leipzig, Leipzig, Germany
| | - Florian Seyfried
- Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Sven Preusser
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Sindy Poppitz
- IFB AdiposityDiseases and.,Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | | | - Konstantinos Gousias
- Department of Neurosurgery, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Jana Hoyer
- Department of Behavioral Epidemiology, Department of Psychology, Technische Universität Dresden, Dresden, Germany
| | | | - Arne Dietrich
- IFB AdiposityDiseases and.,Department of Bariatric Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Karsten Müller
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Burkhard Pleger
- IFB AdiposityDiseases and.,Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.,Collaborative Research Centre 1052 in Obesity Mechanisms, University of Leipzig, Leipzig, Germany.,Department of Neurology and
| |
Collapse
|
41
|
Merlino DJ, Barton JR, Charsar BA, Byrne MD, Rappaport JA, Smeyne RJ, Lepore AC, Snook AE, Waldman SA. Two distinct GUCY2C circuits with PMV (hypothalamic) and SN/VTA (midbrain) origin. Brain Struct Funct 2019; 224:2983-2999. [PMID: 31485718 DOI: 10.1007/s00429-019-01949-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/24/2019] [Indexed: 12/16/2022]
Abstract
Guanylyl cyclase C (GUCY2C) is the afferent central receptor in the gut-brain endocrine axis regulated by the anorexigenic intestinal hormone uroguanylin. GUCY2C mRNA and protein are produced in the hypothalamus, a major center regulating appetite and metabolic homeostasis. Further, GUCY2C mRNA and protein are expressed in the ventral midbrain, a principal structure regulating hedonic reward from behaviors including eating. While GUCY2C is expressed in hypothalamus and midbrain, its precise neuroanatomical organization and relationship with circuits regulating satiety remain unknown. Here, we reveal that hypothalamic GUCY2C mRNA is confined to the ventral premammillary nucleus (PMV), while in midbrain it is produced by neurons in the ventral tegmental area (VTA) and substantia nigra (SN). GUCY2C in the PMV is produced by 46% of neurons expressing anorexigenic leptin receptors, while in the VTA/SN it is produced in most tyrosine hydroxylase-immunoreactive neurons. In contrast to mRNA, GUCY2C protein is widely distributed throughout the brain in canonical sites of PMV and VTA/SN axonal projections. Selective stereotaxic ablation of PMV or VTA/SN neurons eliminated GUCY2C only in their respective canonical projection sites. Conversely, specific anterograde tracer analyses of PMV or VTA/SN neurons confirmed distinct GUCY2C-immunoreactive axons projecting to those canonical locations. Together, these findings reveal two discrete neuronal circuits expressing GUCY2C originating in the PMV in the hypothalamus and in the VTA/SN in midbrain, which separately project to other sites throughout the brain. They suggest a structural basis for a role for the GUCY2C-uroguanylin gut-brain endocrine axis in regulating homeostatic and behavioral components contributing to satiety.
Collapse
Affiliation(s)
- D J Merlino
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, 1020 Locust Street, 368 JAH, Philadelphia, PA, 19107, USA
| | - J R Barton
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, 1020 Locust Street, 368 JAH, Philadelphia, PA, 19107, USA
| | - B A Charsar
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - M D Byrne
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - J A Rappaport
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, 1020 Locust Street, 368 JAH, Philadelphia, PA, 19107, USA
| | - R J Smeyne
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - A C Lepore
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - A E Snook
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, 1020 Locust Street, 368 JAH, Philadelphia, PA, 19107, USA
| | - S A Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, 1020 Locust Street, 368 JAH, Philadelphia, PA, 19107, USA.
| |
Collapse
|
42
|
Liu Y, Yu J, Shi YC, Zhang Y, Lin S. The role of inflammation and endoplasmic reticulum stress in obesity-related cognitive impairment. Life Sci 2019; 233:116707. [PMID: 31374234 DOI: 10.1016/j.lfs.2019.116707] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/16/2019] [Accepted: 07/28/2019] [Indexed: 12/12/2022]
Abstract
The epidemiological investigations and animal model experiments have confirmed the impact of obesity on the brain, behavior, and cognition. However, the mechanism by which obesity affects cognitive function is not fully understood. With the development of an aging society, there is an increase in the economic and social burden caused by the decline in cognitive function. This manuscript reviews the effects of inflammation and endoplasmic reticulum stress (ERS) on the hypothalamus, hippocampus, and the possible impact on cognitive impairment. These findings provide new insights into the pathophysiological mechanisms that lead to the development of cognitive impairment in the context of obesity.
Collapse
Affiliation(s)
- Yilan Liu
- Quanzhou First Hospital, Fujian Medical University, China
| | - Jing Yu
- Quanzhou First Hospital, Fujian Medical University, China
| | - Yan-Chuan Shi
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), China
| | - Yi Zhang
- Quanzhou First Hospital, Fujian Medical University, China.
| | - Shu Lin
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), China; Illawarra Health and Medical Research Institute, Wollongong 2522, Australia.
| |
Collapse
|
43
|
Abstract
Under conditions leading to aging and metabolic syndrome, the hypothalamus atypically undergoes proinflammatory signaling activation leading to a chronic and stable background inflammation, referred to as "hypothalamic microinflammation." Through the past decade of research, progress has been made to causally link this hypothalamic inflammation to the mechanism of aging as well as metabolic syndrome, promoting the "hypothalamic microinflammation" theory, which helps characterize the consensus of these epidemic health problems. In general, it is consistently appreciated that hypothalamic microinflammation emerges during the early stages of aging and metabolic syndrome and evolves to be multifaceted and advanced alongside disease progression, while inhibition of key inflammatory components in the hypothalamus has a broad range of effects in counteracting these disorders. Herein, focusing on aging and metabolic syndrome, this writing aims to provide an overview of and insights into the mediators, signaling components, cellular impacts, and physiological significance of this hypothalamic microinflammation.
Collapse
|
44
|
Rosin JM, Kurrasch DM. Emerging roles for hypothalamic microglia as regulators of physiological homeostasis. Front Neuroendocrinol 2019; 54:100748. [PMID: 31059719 DOI: 10.1016/j.yfrne.2019.100748] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/24/2019] [Accepted: 05/02/2019] [Indexed: 01/22/2023]
Abstract
The hypothalamus is a crucial brain region that responds to external stressors and functions to maintain physiological homeostatic processes, such as core body temperature and energy balance. The hypothalamus regulates homeostasis by producing hormones that thereby influence the production of other hormones that then control the internal milieu of the body. Microglia are resident macrophages and phagocytic immune cells of the central nervous system (CNS), classically known for surveying the brain's environment, responding to neural insults, and disposing of cellular debris. Recent evidence has shown that microglia are also responsive to external stressors and can influence both the development and function of the hypothalamus in a sex-dependent manner. This emerging microglia-hypothalamic interaction raises the intriguing notion that microglia might play an unappreciated role in hypothalamic control of physiological homeostasis. In this review, we briefly outline how the hypothalamus regulates physiological homeostasis and then describe how this literature overlaps with our understanding of microglia's role in the CNS. We also outline the current literature demonstrating how microglia loss or activation affects the hypothalamus, and ultimately homeostasis. We conclude by proposing how microglia could be key regulators of homeostatic processes by sensing cues external to the CNS and transmitting them through the hypothalamus.
Collapse
Affiliation(s)
- Jessica M Rosin
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Deborah M Kurrasch
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
45
|
Radiologic evidence that hypothalamic gliosis is improved after bariatric surgery in obese women with type 2 diabetes. Int J Obes (Lond) 2019; 44:178-185. [PMID: 31201362 PMCID: PMC7366782 DOI: 10.1038/s41366-019-0399-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 04/10/2019] [Accepted: 05/06/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND/OBJECTIVES Hypothalamic neurons play a major role in the control of body mass. Obese subjects present radiologic signs of gliosis in the hypothalamus, which may reflect the damage or loss of neurons involved in whole-body energy homeostasis. It is currently unknown if hypothalamic gliosis (1) differs between obese nondiabetic (ND) and obese diabetic subjects (T2D) or (2) is modified by extensive body mass reduction via Roux-n-Y gastric bypass (RYGB). SUBJECTS/METHODS Fifty-five subjects (all female) including lean controls (CT; n = 13), ND (n = 28), and T2D (n = 14) completed at least one study visit. Subjects underwent anthropometrics and a multi-echo MRI sequence to measure mean bilateral T2 relaxation time in the mediobasal hypothalamus (MBH) and two reference regions (amygdala and putamen). The obese groups underwent RYGB and were re-evaluated 9 months later. Analyses were by linear mixed models. RESULTS Analyses of T2 relaxation time at baseline showed a group by region interaction only in the MBH (P < 0.0001). T2D had longer T2 relaxation times compared to either CT or ND groups. To examine the effects of RYGB on hypothalamic gliosis a three-way (group by region by time) mixed effects model adjusted for age was executed. Group by region (P < 0.0001) and region by time (P = 0.0005) interactions were significant. There was a reduction in MBH relaxation time by RYGB, and, although the T2D group still had higher T2 relaxation time overall compared to the ND group, the T2D group had significantly lower T2 relaxation time after surgery and the ND group showed a trend. The degree of reduction in MBH T2 relaxation time by RYGB was unrelated to clinical outcomes. CONCLUSION T2 relaxation times, a marker of hypothalamic gliosis, are higher in obese women with T2D and are reduced by RYGB-induced weight loss.
Collapse
|
46
|
Chowen JA, Frago LM, Fernández-Alfonso MS. Physiological and pathophysiological roles of hypothalamic astrocytes in metabolism. J Neuroendocrinol 2019; 31:e12671. [PMID: 30561077 DOI: 10.1111/jne.12671] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/27/2018] [Accepted: 12/11/2018] [Indexed: 12/26/2022]
Abstract
The role of glial cells, including astrocytes, in metabolic control has received increasing attention in recent years. Although the original interest in these macroglial cells was a result of astrogliosis being observed in the hypothalamus of diet-induced obese subjects, studies have also focused on how they participate in the physiological control of appetite and energy expenditure. Astrocytes express receptors for numerous hormones, growth factors and neuropeptides. Some functions of astrocytes include transport of nutrients and hormones from the circulation to the brain, storage of glycogen, participation in glucose sensing, synaptic plasticity, uptake and metabolism of neurotransmitters, release of substances to modify neurotransmission, and cytokine production, amongst others. In the hypothalamus, these physiological glial functions impact on neuronal circuits that control systemic metabolism to modify their outputs. The initial response of astrocytes to poor dietary habits and obesity involves activation of neuroprotective mechanisms but, with chronic exposure to these situations, hypothalamic astrocytes participate in the development of some of the damaging secondary effects. The present review discusses not only some of the physiological functions of hypothalamic astrocytes in metabolism, but also their role in the secondary complications of obesity, such as insulin resistance and cardiovascular affectations.
Collapse
Affiliation(s)
- Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Madrid, Spain
- CIBEROBN (Centro de Investigación Biomédica en Red sobre Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
| | - Laura M Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Madrid, Spain
- CIBEROBN (Centro de Investigación Biomédica en Red sobre Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
| | - María Soledad Fernández-Alfonso
- Instituto Pluridisciplinar UCM y Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| |
Collapse
|
47
|
MacDonald AJ, Robb JL, Morrissey NA, Beall C, Ellacott KLJ. Astrocytes in neuroendocrine systems: An overview. J Neuroendocrinol 2019; 31:e12726. [PMID: 31050045 DOI: 10.1111/jne.12726] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/26/2019] [Accepted: 04/28/2019] [Indexed: 12/11/2022]
Abstract
A class of glial cell, astrocytes, is highly abundant in the central nervous system (CNS). In addition to maintaining tissue homeostasis, astrocytes regulate neuronal communication and synaptic plasticity. There is an ever-increasing appreciation that astrocytes are involved in the regulation of physiology and behaviour in normal and pathological states, including within neuroendocrine systems. Indeed, astrocytes are direct targets of hormone action in the CNS, via receptors expressed on their surface, and are also a source of regulatory neuropeptides, neurotransmitters and gliotransmitters. Furthermore, as part of the neurovascular unit, astrocytes can regulate hormone entry into the CNS. This review is intended to provide an overview of how astrocytes are impacted by and contribute to the regulation of a diverse range of neuroendocrine systems: energy homeostasis and metabolism, reproduction, fluid homeostasis, the stress response and circadian rhythms.
Collapse
Affiliation(s)
- Alastair J MacDonald
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Josephine L Robb
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Nicole A Morrissey
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Craig Beall
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Kate L J Ellacott
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| |
Collapse
|
48
|
Macedo F, Dos Santos LS, Glezer I, da Cunha FM. Brain Innate Immune Response in Diet-Induced Obesity as a Paradigm for Metabolic Influence on Inflammatory Signaling. Front Neurosci 2019; 13:342. [PMID: 31068773 PMCID: PMC6491681 DOI: 10.3389/fnins.2019.00342] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/25/2019] [Indexed: 12/19/2022] Open
Abstract
Obesity is a predisposing factor for numerous morbidities, including those affecting the central nervous system. Hypothalamic inflammation is a hallmark of obesity and is believed to participate in the onset and progression of the obese phenotype, by promoting changes in neuronal functions involved in the control of metabolism. The activation of brain immune cells in the hypothalamus, which are represented by microglia and brain macrophages, is associated with obesity and has been the focus of intense research. Despite the significant body of knowledge gathered on this topic, obesity-induced metabolic changes in brain cells involved in innate immune responses are still poorly characterized due, at least in part, to limitations in the existing experimental methods. Since the metabolic state influences immune responses of microglia and other myeloid cells, the understanding and characterization of the effects of cellular metabolism on the functions of these cells, and their impact on brain integrity, are crucial for the development of efficient therapeutic interventions for individuals exposed to a long-term high fat diet (HFD). Here we review and speculate on the cellular basis that may underlie the observed changes in the reactivity and metabolism of the innate immune cells of the brain in diet-induced obesity (DIO), and discuss important points that deserve further investigation.
Collapse
Affiliation(s)
- Felipe Macedo
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Lucas Souza Dos Santos
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Isaias Glezer
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fernanda Marques da Cunha
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
49
|
Harrison L, Pfuhlmann K, Schriever SC, Pfluger PT. Profound weight loss induces reactive astrogliosis in the arcuate nucleus of obese mice. Mol Metab 2019; 24:149-155. [PMID: 30979678 PMCID: PMC6977167 DOI: 10.1016/j.molmet.2019.03.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 03/27/2019] [Accepted: 03/28/2019] [Indexed: 11/12/2022] Open
Abstract
Objective Obesity has been linked to an inflammation like state in the hypothalamus, mainly characterized by reactive gliosis (RG) of astrocytes and microglia. Here, using two diet models or pharmacological treatment, we assessed the effects of mild and drastic weight loss on RG, in the context of high-fat diet (HFD) induced obesity. Methods We subjected HFD-induced obese (DIO) male C57BL/6J mice to a weight loss intervention with a switch to standard chow, calorie restriction (CR), or treatment with the Glp1 receptor agonist Exendin-4 (EX4). The severity of RG was estimated by an ordinal scoring system based on fluorescence intensities of glial fibrillary acidic protein, ionized calcium-binding adapter molecule 1 positive (Iba1), cell numbers, and morphological characteristics. Results In contrast to previous reports, DIO mice fed chronically with HFD showed no differences in microglial or astrocytic RG, compared to chow controls. Moreover, mild or profound weight loss had no impact on microglial RG. However, astrocyte RG was increased in CR and EX4 groups compared to chow fed animals and strongly correlated to body weight loss. Profound weight loss by either CR or EX4 was further linked to increased levels of circulating non-esterified free fatty acids. Conclusions Overall, our data demonstrate that in a chronically obese state, astrocyte and microglial RG is indifferent from that observed in age-matched chow controls. Nonetheless, profound acute weight loss can induce astrocyte RG in the hypothalamic arcuate nucleus, possibly due to increased circulating NEFAs. This suggests that astrocytes may sense acute changes to both the dietary environment and body weight. Acute weight loss induces reactive gliosis in arcuate nucleus residing astrocytes. Reactive gliosis after chronic high-fat diet is comparable to chow fed litter mates. Observed gliosis coincides with increased circulating non-esterified fatty acids.
Collapse
Affiliation(s)
- Luke Harrison
- Research Unit Neurobiology of Diabetes, Helmholtz Zentrum München, 85764, Neuherberg, Germany; Institute for Diabetes and Obesity, Helmholtz Zentrum München, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany; Division of Metabolic Diseases, Technische Universität München, 80333, Munich, Germany
| | - Katrin Pfuhlmann
- Research Unit Neurobiology of Diabetes, Helmholtz Zentrum München, 85764, Neuherberg, Germany; Institute for Diabetes and Obesity, Helmholtz Zentrum München, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany; Division of Metabolic Diseases, Technische Universität München, 80333, Munich, Germany
| | - Sonja C Schriever
- Research Unit Neurobiology of Diabetes, Helmholtz Zentrum München, 85764, Neuherberg, Germany; Institute for Diabetes and Obesity, Helmholtz Zentrum München, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Paul T Pfluger
- Research Unit Neurobiology of Diabetes, Helmholtz Zentrum München, 85764, Neuherberg, Germany; Institute for Diabetes and Obesity, Helmholtz Zentrum München, 85764, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany.
| |
Collapse
|
50
|
Chen D, Cao S, Chang B, Ma T, Gao H, Tong Y, Li T, Han J, Yi X. Increasing hypothalamic nucleobindin 2 levels and decreasing hypothalamic inflammation in obese male mice via diet and exercise alleviate obesity-associated hypogonadism. Neuropeptides 2019; 74:34-43. [PMID: 30503692 DOI: 10.1016/j.npep.2018.10.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/26/2018] [Accepted: 10/30/2018] [Indexed: 02/08/2023]
Abstract
To explore the role of nesfatin-1 in regulating male reproductive function during energy balance variation, we employed an obese mouse model which was first induced by a high-fat diet (HFD) and followed by interventions of a normal diet (ND) and/or moderate exercise, and then serum reproductive hormones of male mice, hypothalamic nucleobindin 2 (NUCB2)/nesfatin-1, inflammatory factors, and gonadotropin-releasing hormone (GnRH) levels were tested. Our findings showed that both serum nesfatin-1, follicle-stimulating hormone (FSH), luteinizing hormone (LH), and testosterone (T) levels and hypothalamic NUCB2/nesfatin-1 and Gnrh mRNA levels were reduced, whereas, the mRNA and protein levels of hypothalamic tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, inhibitor kappa B kinase β (IKKβ), and nuclear factor (NF)-κB were increased in obese male mice. Diet, exercise, and diet combined with exercise interventions reversed the decreases in serum nesfatin-1, FSH, LH, and T levels; increased hypothalamic NUCB2/nesfatin-1 and Gnrh mRNA levels; and reduced hypothalamic TNF-α, IL-1β, IKKβ, and NF-κB levels. These changes were accompanied by reduced adiposity, and these effects were more obvious in the diet combined with exercise group. Overall, our findings suggested that the hypogonadotropic hypogonadism associated with obesity may be induced by reduced hypothalamic NUCB2/nesfatin-1 levels, which attenuated the stimulatory effect on GnRH directly or indirectly by suppressing its anti-inflammatory effect in the brain. Diet and/or exercise interventions were able to alleviate the hypogonadotropic hypogonadism associated with obesity, potentially by increasing hypothalamic NUCB2/nesfatin-1 levels.
Collapse
Affiliation(s)
- Dequan Chen
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, PR China; School of Physical Education, Minnan Normal Universtiy, Zhangzhou, Fujian 363000, PR China
| | - Shicheng Cao
- Department of Sport Medicine, School of Fundamental Sciences, China Medical University, Shenyang, Liaoning 110001, PR China
| | - Bo Chang
- School of Kinesiology, Shenyang Sport University, Shenyang, Liaoning 110102, PR China
| | - Tie Ma
- School of Kinesiology, Shenyang Sport University, Shenyang, Liaoning 110102, PR China
| | - Haining Gao
- School of Kinesiology, Shenyang Sport University, Shenyang, Liaoning 110102, PR China
| | - Yao Tong
- School of Kinesiology, Shenyang Sport University, Shenyang, Liaoning 110102, PR China
| | - Tao Li
- School of Kinesiology, Shenyang Sport University, Shenyang, Liaoning 110102, PR China
| | - Junchao Han
- School of Kinesiology, Shenyang Sport University, Shenyang, Liaoning 110102, PR China
| | - Xuejie Yi
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, PR China; School of Kinesiology, Shenyang Sport University, Shenyang, Liaoning 110102, PR China.
| |
Collapse
|