1
|
Sanchez C, Nadal M, Cansell C, Laroui S, Descombes X, Rovère C, Debreuve É. Computational detection, characterization, and clustering of microglial cells in a mouse model of fat-induced postprandial hypothalamic inflammation. Methods 2025; 236:28-38. [PMID: 40021035 DOI: 10.1016/j.ymeth.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025] Open
Abstract
Obesity is associated with brain inflammation, glial reactivity, and immune cells infiltration. Studies in rodents have shown that glial reactivity occurs within 24 h of high-fat diet (HFD) consumption, long before obesity development, and takes place mainly in the hypothalamus (HT), a crucial brain structure for controlling body weight. Understanding more precisely the kinetics of glial activation of two major brain cells (astrocytes and microglia) and their impact on eating behavior could prevent obesity and offer new prospects for therapeutic treatments. To understand the mechanisms pertaining to obesity-related neuroinflammation, we developed a fully automated algorithm, NutriMorph. Although some algorithms were developed in the past decade to detect and segment cells, they are highly specific, not fully automatic, and do not provide the desired morphological analysis. Our algorithm copes with these issues and performs the analysis of cells images (here, microglia of the hypothalamic arcuate nucleus), and the morphological clustering of these cells through statistical analysis and machine learning. Using the k-Means algorithm, it clusters the microglia of the control condition (healthy mice) and the different states of neuroinflammation induced by high-fat diets (obese mice) into subpopulations. This paper is an extension and re-analysis of a first published paper showing that microglial reactivity can already be seen after few hours of high-fat diet (Cansell et al., 2021 [5]). Thanks to NutriMorph algorithm, we unravel the presence of different hypothalamic microglial subpopulations (based on morphology) subject to proportion changes in response to already few hours of high-fat diet in mice.
Collapse
Affiliation(s)
- Clara Sanchez
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France
| | - Morgane Nadal
- Université Côte d'Azur, CNRS, Inria, I3S, Team Morpheme, Sophia Antipolis, France
| | | | - Sarah Laroui
- Université Côte d'Azur, CNRS, Inria, I3S, Team Morpheme, Sophia Antipolis, France
| | - Xavier Descombes
- Université Côte d'Azur, CNRS, Inria, I3S, Team Morpheme, Sophia Antipolis, France
| | - Carole Rovère
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France
| | - Éric Debreuve
- Université Côte d'Azur, CNRS, Inria, I3S, Team Morpheme, Sophia Antipolis, France.
| |
Collapse
|
2
|
Sharif A, Prevot V. Astrogenesis in the hypothalamus: A life-long process contributing to the development and plasticity of neuroendocrine networks. Front Neuroendocrinol 2024; 75:101154. [PMID: 39226950 DOI: 10.1016/j.yfrne.2024.101154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024]
Abstract
Astrocytes are now recognized as integral components of neural circuits, regulating their maturation, activity and plasticity. Neuroendocrinology has provided fertile ground for revealing the diverse strategies used by astrocytes to regulate the physiological and behavioural outcomes of neural circuit activity in response to internal and environmental inputs. However, the development of astrocytes in the hypothalamus has received much less attention than in other brain regions such as the cerebral cortex and spinal cord. In this review, we synthesize our current knowledge of astrogenesis in the hypothalamus across various life stages. A distinctive feature of hypothalamic astrogenesis is that it persists all throughout lifespan, and involves multiple cellular sources corresponding to radial glial cells during early development, followed by tanycytes, parenchymal progenitors and locally dividing astrocytes. Astrogenesis in the hypothalamus is closely coordinated with the maturation of hypothalamic neurons. This coordination is exemplified by recent findings in neurons producing gonadotropin-releasing hormone, which actively shape their astroglial environment during infancy to integrate functionally into their neural network and facilitate sexual maturation, a process vulnerable to endocrine disruption. While hypothalamic astrogenesis shares common principles with other brain regions, it also exhibits specific features in its dynamics and regulation, both at the inter- and intra-regional levels. These unique properties emphasize the importance of further exploration. Additionally, we discuss the experimental strategies used to assess astrogenesis in the hypothalamus and their potential bias and limitations. Understanding the mechanisms of hypothalamic astrogenesis throughout life will be crucial for comprehending the development and function of the hypothalamus under both physiological and pathological conditions.
Collapse
Affiliation(s)
- Ariane Sharif
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, FHU 1000 Days for Health, Lille, France.
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, FHU 1000 Days for Health, Lille, France.
| |
Collapse
|
3
|
Barros LF, Schirmeier S, Weber B. The Astrocyte: Metabolic Hub of the Brain. Cold Spring Harb Perspect Biol 2024; 16:a041355. [PMID: 38438188 PMCID: PMC11368191 DOI: 10.1101/cshperspect.a041355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Astrocytic metabolism has taken center stage. Interposed between the neuron and the vasculature, astrocytes exert control over the fluxes of energy and building blocks required for neuronal activity and plasticity. They are also key to local detoxification and waste recycling. Whereas neurons are metabolically rigid, astrocytes can switch between different metabolic profiles according to local demand and the nutritional state of the organism. Their metabolic state even seems to be instructive for peripheral nutrient mobilization and has been implicated in information processing and behavior. Here, we summarize recent progress in our understanding of astrocytic metabolism and its effects on metabolic homeostasis and cognition.
Collapse
Affiliation(s)
- L Felipe Barros
- Centro de Estudios Científicos, Valdivia 5110465, Chile
- Universidad San Sebastián, Facultad de Medicina y Ciencia, Valdivia 5110693, Chile
| | - Stefanie Schirmeier
- Technische Universität Dresden, Department of Biology, 01217 Dresden, Germany
| | - Bruno Weber
- University of Zurich, Institute of Pharmacology and Toxicology, 8057 Zurich, Switzerland
| |
Collapse
|
4
|
Srour N, Caron A, Michael NJ. Do POMC neurons have a sweet tooth for leptin? Special issue: Role of nutrients in nervous control of energy balance. Biochimie 2024; 223:179-187. [PMID: 36122808 DOI: 10.1016/j.biochi.2022.09.006] [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: 05/21/2022] [Revised: 08/29/2022] [Accepted: 09/09/2022] [Indexed: 11/19/2022]
Abstract
Coordinated detection of changes in metabolic state by the nervous system is fundamental for survival. Hypothalamic pro-opiomelanocortin (POMC) neurons play a critical role in integrating metabolic signals, including leptin levels. They also coordinate adaptative responses and thus represent an important relay in the regulation of energy balance. Despite a plethora of work documenting the effects of individual hormones, nutrients, and neuropeptides on POMC neurons, the importance for crosstalk and additive effects between such signaling molecules is still underexplored. The ability of the metabolic state and the concentrations of nutrients, such as glucose, to influence leptin's effects on POMC neurons appears critical for understanding the function and complexity of this regulatory network. Here, we summarize the current knowledge on the effects of leptin on POMC neuron electrical excitability and discuss factors potentially contributing to variability in these effects, with a particular focus on the mouse models that have been developed and the importance of extracellular glucose levels. This review highlights the importance of the metabolic "environment" for determining hypothalamic neuronal responsiveness to metabolic cues and for determining the fundamental effects of leptin on the activity of hypothalamic POMC neurons.
Collapse
Affiliation(s)
- Nader Srour
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada; Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Alexandre Caron
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada; Faculté de Pharmacie, Université Laval, Québec, QC, Canada; Montreal Diabetes Research Center, QC, Canada.
| | - Natalie Jane Michael
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada; Faculté de Pharmacie, Université Laval, Québec, QC, Canada.
| |
Collapse
|
5
|
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
|
6
|
Diao S, Chen C, Benani A, Magnan C, Van Steenwinckel J, Gressens P, Cruciani-Guglielmacci C, Jacquens A, Bokobza C. Preterm birth: A neuroinflammatory origin for metabolic diseases? Brain Behav Immun Health 2024; 37:100745. [PMID: 38511150 PMCID: PMC10950814 DOI: 10.1016/j.bbih.2024.100745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/16/2024] [Accepted: 02/21/2024] [Indexed: 03/22/2024] Open
Abstract
Preterm birth and its related complications have become more and more common as neonatal medicine advances. The concept of "developmental origins of health and disease" has raised awareness of adverse perinatal events in the development of diseases later in life. To explore this concept, we propose that encephalopathy of prematurity (EoP) as a potential pro-inflammatory early life event becomes a novel risk factor for metabolic diseases in children/adolescents and adulthood. Here, we review epidemiological evidence that links preterm birth to metabolic diseases and discuss possible synergic roles of preterm birth and neuroinflammation from EoP in the development of metabolic diseases. In addition, we explore theoretical underlying mechanisms regarding developmental programming of the energy control system and HPA axis.
Collapse
Affiliation(s)
- Sihao Diao
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, 201102, China
- Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Chao Chen
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, 201102, China
- Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Alexandre Benani
- CSGA, Centre des Sciences du Goût et de l'Alimentation, UMR 6265 CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | | | | | - Pierre Gressens
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| | | | - Alice Jacquens
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
- Department of Anesthesia and Critical Care, APHP-Sorbonne University, Hôpital La Pitié- Salpêtrière, Paris, France
| | - Cindy Bokobza
- Université Paris Cité, Inserm, NeuroDiderot, 75019, Paris, France
| |
Collapse
|
7
|
Ameroso D, Rios M. Synaptic plasticity and the role of astrocytes in central metabolic circuits. WIREs Mech Dis 2024; 16:e1632. [PMID: 37833830 PMCID: PMC10842964 DOI: 10.1002/wsbm.1632] [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: 07/01/2023] [Revised: 09/08/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023]
Abstract
Neural circuits in the brain, primarily in the hypothalamus, are paramount to the homeostatic control of feeding and energy utilization. They integrate hunger, satiety, and body adiposity cues from the periphery and mediate the appropriate behavioral and physiological responses to satisfy the energy demands of the animal. Notably, perturbations in central homeostatic circuits have been linked to the etiology of excessive feeding and obesity. Considering the ever-changing energy requirements of the animal and required adaptations, it is not surprising that brain-feeding circuits remain plastic in adulthood and are subject to changes in synaptic strength as a consequence of nutritional status. Indeed, synapse density, probability of presynaptic transmitter release, and postsynaptic responses in hypothalamic energy balance centers are tailored to behavioral and physiological responses required to sustain survival. Mounting evidence supports key roles of astrocytes facilitating some of this plasticity. Here we discuss these synaptic plasticity mechanisms and the emerging roles of astrocytes influencing energy and glucose balance control in health and disease. This article is categorized under: Cancer > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Dominique Ameroso
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
| | - Maribel Rios
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111
| |
Collapse
|
8
|
Ko HG, Chun H, Han S, Kaang BK. Role of spinal astrocytes through the perisynaptic astrocytic process in pathological pain. Mol Brain 2023; 16:81. [PMID: 38093330 PMCID: PMC10717263 DOI: 10.1186/s13041-023-01069-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/07/2023] [Indexed: 12/17/2023] Open
Abstract
Pathological pain is caused by abnormal activity in the neural circuit that transmits nociceptive stimuli. Beyond homeostatic functions, astrocytes actively participate in regulating synaptic transmission as members of tripartite synapses. The perisynaptic astrocytic process (PAP) is the key structure that allows astrocytes to play these roles and not only physically supports synapse formation through cell adhesion molecules (CAMs) but also regulates the efficiency of chemical signaling. Accumulating evidence has revealed that spinal astrocytes are involved in pathological pain by modulating the efficacy of neurotransmitters such as glutamate and GABA through transporters located in the PAP and by directly regulating synaptic transmission through various gliotransmitters. Although various CAMs contribute to pathological pain, insufficient evidence is available as to whether astrocytic CAMs also have this role. Therefore, more in-depth research is needed on how pathological pain is induced and maintained by astrocytes, especially in the PAP surrounding the synapse, and this will subsequently increase our understanding and treatment of pathological pain.
Collapse
Affiliation(s)
- Hyoung-Gon Ko
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, 2177 Dalgubeol- daero, Daegu, 41940, South Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, South Korea
| | - Heejung Chun
- College of Pharmacy, Yonsei-SL Bigen Institute (YSLI), Yonsei University, Incheon, South Korea
| | - Seunghyo Han
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, 2177 Dalgubeol- daero, Daegu, 41940, South Korea
| | - Bong-Kiun Kaang
- Center for Cognition and Sociality, Life Science Institute, Institute for Basic Science (IBS), Daejeon, 34141, South Korea.
| |
Collapse
|
9
|
Meng A, Ameroso D, Rios M. mGluR5 in Astrocytes in the Ventromedial Hypothalamus Regulates Pituitary Adenylate Cyclase-Activating Polypeptide Neurons and Glucose Homeostasis. J Neurosci 2023; 43:5918-5935. [PMID: 37507231 PMCID: PMC10436691 DOI: 10.1523/jneurosci.0193-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/09/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023] Open
Abstract
The ventromedial hypothalamus (VMH) is a functionally heterogeneous nucleus critical for systemic energy, glucose, and lipid balance. We showed previously that the metabotropic glutamate receptor 5 (mGluR5) plays essential roles regulating excitatory and inhibitory transmission in SF1+ neurons of the VMH and facilitating glucose and lipid homeostasis in female mice. Although mGluR5 is also highly expressed in VMH astrocytes in the mature brain, its role there influencing central metabolic circuits is unknown. In contrast to the glucose intolerance observed only in female mice lacking mGluR5 in VMH SF1 neurons, selective depletion of mGluR5 in VMH astrocytes enhanced glucose tolerance without affecting food intake or body weight in both adult female and male mice. The improved glucose tolerance was associated with elevated glucose-stimulated insulin release. Astrocytic mGluR5 male and female mutants also exhibited reduced adipocyte size and increased sympathetic tone in gonadal white adipose tissue. Diminished excitatory drive and synaptic inputs onto VMH Pituitary adenylate cyclase-activating polypeptide (PACAP+) neurons and reduced activity of these cells during acute hyperglycemia underlie the observed changes in glycemic control. These studies reveal an essential role of astrocytic mGluR5 in the VMH regulating the excitatory drive onto PACAP+ neurons and activity of these cells facilitating glucose homeostasis in male and female mice.SIGNIFICANCE STATEMENT Neuronal circuits within the VMH play chief roles in the regulation of whole-body metabolic homeostasis. It remains unclear how astrocytes influence neurotransmission in this region to facilitate energy and glucose balance control. Here, we explored the role of the metabotropic glutamate receptor, mGluR5, using a mouse model with selective depletion of mGluR5 from VMH astrocytes. We show that astrocytic mGluR5 critically regulates the excitatory drive and activity of PACAP-expressing neurons in the VMH to control glucose homeostasis in both female and male mice. Furthermore, mGluR5 in VMH astrocytes influences adipocyte size and sympathetic tone in white adipose tissue. These studies provide novel insight toward the importance of hypothalamic astrocytes participating in central circuits regulating peripheral metabolism.
Collapse
Affiliation(s)
- Alice Meng
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Dominique Ameroso
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts 02111, United States
| | - Maribel Rios
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts 02111
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts 02111, United States
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| |
Collapse
|
10
|
MacDonald AJ, Pye KR, Beall C, Ellacott KLJ. Impact of chemogenetic activation of dorsal vagal complex astrocytes in mice on adaptive glucoregulatory responses. J Neuroendocrinol 2023; 35:e13315. [PMID: 37370201 DOI: 10.1111/jne.13315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 05/13/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023]
Abstract
The dorsal vagal complex (DVC) regulates diverse aspects of physiology including food intake and blood glucose homeostasis. Astrocytes play an active role in regulating DVC function and, by extension, physiological parameters. DVC astrocytes in ex vivo slices respond to low tissue glucose. The response of neurons to low glucose is conditional on intact astrocyte signalling in slice preparations, suggesting astrocytes are primary sensors of glucose deprivation (glucoprivation). Based on these published findings we hypothesised that in vivo DVC astrocyte manipulation with chemogenetics would be sufficient to alter physiological responses that control blood glucose. We found that 2-h after systemic 2-DG-induced glucoprivation there were no observable changes in morphology of glial fibrillary acidic protein (GFAP)-immunoreactive DVC cells, specifically those in the nucleus of the solitary tract (NTS). Chemogenetic activation of DVC astrocytes was sufficient to suppress nocturnal food intake by reducing both meal size and meal number and this manipulation also suppressed 2-DG-induced glucoprivic food intake. Chemogenetic activation of DVC astrocytes did not increase basal blood glucose nor protect against insulin-induced hypoglycaemia. In male mice, chemogenetic DVC astrocyte activation did not alter glucose tolerance. In female mice, the initial glucose excursion was reduced in a glucose tolerance test, suggesting enhanced glucose absorption. Based on our data and published work, we propose that DVC astrocytes may play an indispensable homeostatic role, that is, are necessary to maintain the function of glucoregulatory neuronal circuitry, but alone their bulk activation is not sufficient to result in adaptive glucoregulatory responses. It is possible that there are state-dependent effects and/or DVC astrocyte subsets that have this specialised role, but this was unresolvable using the experimental approaches employed here.
Collapse
Affiliation(s)
- Alastair J MacDonald
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Katherine R Pye
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Craig Beall
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Kate L J Ellacott
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, UK
| |
Collapse
|
11
|
Haspula D, Cui Z. Neurochemical Basis of Inter-Organ Crosstalk in Health and Obesity: Focus on the Hypothalamus and the Brainstem. Cells 2023; 12:1801. [PMID: 37443835 PMCID: PMC10341274 DOI: 10.3390/cells12131801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/23/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Precise neural regulation is required for maintenance of energy homeostasis. Essential to this are the hypothalamic and brainstem nuclei which are located adjacent and supra-adjacent to the circumventricular organs. They comprise multiple distinct neuronal populations which receive inputs not only from other brain regions, but also from circulating signals such as hormones, nutrients, metabolites and postprandial signals. Hence, they are ideally placed to exert a multi-tier control over metabolism. The neuronal sub-populations present in these key metabolically relevant nuclei regulate various facets of energy balance which includes appetite/satiety control, substrate utilization by peripheral organs and glucose homeostasis. In situations of heightened energy demand or excess, they maintain energy homeostasis by restoring the balance between energy intake and expenditure. While research on the metabolic role of the central nervous system has progressed rapidly, the neural circuitry and molecular mechanisms involved in regulating distinct metabolic functions have only gained traction in the last few decades. The focus of this review is to provide an updated summary of the mechanisms by which the various neuronal subpopulations, mainly located in the hypothalamus and the brainstem, regulate key metabolic functions.
Collapse
Affiliation(s)
- Dhanush Haspula
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Zhenzhong Cui
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA;
| |
Collapse
|
12
|
Vaccari-Cardoso B, Antipina M, Teschemacher AG, Kasparov S. Lactate-Mediated Signaling in the Brain-An Update. Brain Sci 2022; 13:brainsci13010049. [PMID: 36672031 PMCID: PMC9856103 DOI: 10.3390/brainsci13010049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/15/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Lactate is a universal metabolite produced and released by all cells in the body. Traditionally it was viewed as energy currency that is generated from pyruvate at the end of the glycolytic pathway and sent into the extracellular space for other cells to take up and consume. In the brain, such a mechanism was postulated to operate between astrocytes and neurons many years ago. Later, the discovery of lactate receptors opened yet another chapter in the quest to understand lactate actions. Other ideas, such as modulation of NMDA receptors were also proposed. Up to this day, we still do not have a consensus view on the relevance of any of these mechanisms to brain functions or their contribution to human or animal physiology. While the field develops new ideas, in this brief review we analyze some recently published studies in order to focus on some unresolved controversies and highlight the limitations that need to be addressed in future work. Clearly, only by using similar and overlapping methods, cross-referencing experiments, and perhaps collaborative efforts, we can finally understand what the role of lactate in the brain is and why this ubiquitous molecule is so important.
Collapse
Affiliation(s)
- Barbara Vaccari-Cardoso
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| | - Maria Antipina
- MEDBIO, Immanuel Kant Baltic Federal University, Universitetskaya Str., 2, 236041 Kaliningrad, Russia
| | - Anja G. Teschemacher
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| | - Sergey Kasparov
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK
- Correspondence:
| |
Collapse
|
13
|
Bouyakdan K, Manceau R, Robb JL, Rodaros D, Fulton S, Alquier T. Role of astroglial ACBP in energy metabolism flexibility and feeding responses to metabolic challenges in male mice. J Neuroendocrinol 2022; 34:e13218. [PMID: 36471907 DOI: 10.1111/jne.13218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/26/2022] [Accepted: 11/10/2022] [Indexed: 11/19/2022]
Abstract
Acyl-CoA binding protein (ACBP), also known as diazepam binding inhibitor (DBI), has recently emerged as a hypothalamic and brainstem gliopeptide regulating energy balance. Previous work has shown that the ACBP-derived octadecaneuropeptide exerts strong anorectic action via proopiomelanocortin (POMC) neuron activation and the melanocortin-4 receptor. Importantly, targeted ACBP loss-of-function in astrocytes promotes hyperphagia and diet-induced obesity while its overexpression in arcuate astrocytes reduces feeding and body weight. Despite this knowledge, the role of astroglial ACBP in adaptive feeding and metabolic responses to acute metabolic challenges has not been investigated. Using different paradigms, we found that ACBP deletion in glial fibrillary acidic protein (GFAP)-positive astrocytes does not affect weight loss when obese male mice are transitioned from a high fat diet to a chow diet, nor metabolic parameters in mice fed with a normal chow diet (e.g., energy expenditure, body temperature) during fasting, cold exposure and at thermoneutrality. In contrast, astroglial ACBP deletion impairs meal pattern and feeding responses during refeeding after a fast and during cold exposure, thereby showing that ACBP is required to stimulate feeding in states of increased energy demand. These findings challenge the general view that astroglial ACBP exerts anorectic effects and suggest that regulation of feeding by ACBP is dependent on metabolic status.
Collapse
Affiliation(s)
- Khalil Bouyakdan
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal Diabetes Research Center, and Departments of Medicine and Neurosciences and Nutrition, Université de Montréal, Montréal, Quebec, Canada
| | - Romane Manceau
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal Diabetes Research Center, and Departments of Medicine and Neurosciences and Nutrition, Université de Montréal, Montréal, Quebec, Canada
| | - Josephine L Robb
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal Diabetes Research Center, and Departments of Medicine and Neurosciences and Nutrition, Université de Montréal, Montréal, Quebec, Canada
| | - Demetra Rodaros
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal Diabetes Research Center, and Departments of Medicine and Neurosciences and Nutrition, Université de Montréal, Montréal, Quebec, Canada
| | - Stephanie Fulton
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal Diabetes Research Center, and Departments of Medicine and Neurosciences and Nutrition, Université de Montréal, Montréal, Quebec, Canada
| | - Thierry Alquier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal Diabetes Research Center, and Departments of Medicine and Neurosciences and Nutrition, Université de Montréal, Montréal, Quebec, Canada
| |
Collapse
|
14
|
Campillo BW, Galguera D, Cerdan S, López-Larrubia P, Lizarbe B. Short-term high-fat diet alters the mouse brain magnetic resonance imaging parameters consistently with neuroinflammation on males and metabolic rearrangements on females. A pre-clinical study with an optimized selection of linear mixed-effects models. Front Neurosci 2022; 16:1025108. [PMID: 36507349 PMCID: PMC9729798 DOI: 10.3389/fnins.2022.1025108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/20/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction High-fat diet (HFD) consumption is known to trigger an inflammatory response in the brain that prompts the dysregulation of energy balance, leads to insulin and leptin resistance, and ultimately obesity. Obesity, at the same, has been related to cerebral magnetic resonance imaging (MRI) alterations, but the onset of HFD-induced neuroinflammation, however, has been principally reported on male rodents and by ex vivo methods, with the effects on females and the origin of MRI changes remaining unassessed. Methods We characterized the onset and evolution of obesity on male and female mice during standard or HFD administration by physiological markers and multiparametric MRI on four cerebral regions involved in appetite regulation and energy homeostasis. We investigated the effects of diet, time under diet, brain region and sex by identifying their significant contributions to sequential linear mixed-effects models, and obtained their regional neurochemical profiles by high-resolution magic angle spinning spectroscopy. Results Male mice developed an obese phenotype paralleled by fast increases in magnetization transfer ratio values, while females delayed the obesity progress and showed no MRI-signs of cerebral inflammation, but larger metabolic rearrangements on the neurochemical profile. Discussion Our study reveals early MRI-detectable changes compatible with the development of HFD-induced cerebral cytotoxic inflammation on males but suggest the existence of compensatory metabolic adaptations on females that preclude the corresponding detection of MRI alterations.
Collapse
Affiliation(s)
- Basilio Willem Campillo
- Instituto de Investigaciones Biomédicas Alberto Sols (IIBm), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - David Galguera
- Instituto de Investigaciones Biomédicas Alberto Sols (IIBm), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Sebastian Cerdan
- Instituto de Investigaciones Biomédicas Alberto Sols (IIBm), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Pilar López-Larrubia
- Instituto de Investigaciones Biomédicas Alberto Sols (IIBm), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain,Pilar López-Larrubia,
| | - Blanca Lizarbe
- Instituto de Investigaciones Biomédicas Alberto Sols (IIBm), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain,Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain,*Correspondence: Blanca Lizarbe,
| |
Collapse
|
15
|
He Z, Xu Y, Ma Q, Zhou C, Yang L, Lin M, Deng P, Yang Z, Gong M, Zhang H, Lu M, Li Y, Gao P, Lu Y, He M, Zhang L, Pi H, Zhang K, Qin S, Yu Z, Zhou Z, Chen C. SOX2 modulated astrocytic process plasticity is involved in arsenic-induced metabolic disorders. JOURNAL OF HAZARDOUS MATERIALS 2022; 435:128942. [PMID: 35468398 DOI: 10.1016/j.jhazmat.2022.128942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/02/2022] [Accepted: 04/13/2022] [Indexed: 06/14/2023]
Abstract
Metabolic disorders induced by arsenic exposure have attracted great public concern. However, it remains unclear whether hypothalamus-based central regulation mechanisms are involved in this process. Here, we exposed mice to 100 μg/L arsenic in drinking water and established a chronic arsenic exposure model. Our study revealed that chronic arsenic exposure caused metabolic disorders in mice including impaired glucose metabolism and decreased energy expenditure. Arsenic exposure also impaired glucose sensing and the activation of proopiomelanocortin (POMC) neurons in the hypothalamus. In particular, arsenic exposure damaged the plasticity of hypothalamic astrocytic process. Further research revealed that arsenic exposure inhibited the expression of sex-determining region Y-Box 2 (SOX2), which decreased the expression level of insulin receptors (INSRs) and the phosphorylation of AKT. The conditional deletion of astrocytic SOX2 exacerbated arsenic-induced effects on metabolic disorders, the impairment of hypothalamic astrocytic processes, and the inhibition of INSR/AKT signaling. Furthermore, the arsenic-induced impairment of astrocytic processes and inhibitory effects on INSR/AKT signaling were reversed by SOX2 overexpression in primary hypothalamic astrocytes. Together, we demonstrated here that chronic arsenic exposure caused metabolic disorders by impairing SOX2-modulated hypothalamic astrocytic process plasticity in mice. Our study provides evidence of novel central regulatory mechanisms underlying arsenic-induced metabolic disorders and emphasizes the crucial role of SOX2 in regulating the process plasticity of adult astrocytes.
Collapse
Affiliation(s)
- Zhixin He
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Yudong Xu
- Department of Environmental Medicine, School of Public Health, and Department of Emergency Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Qinlong Ma
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Chao Zhou
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China; Army 953 Hospital, Shigatse Branch of Xinqiao Hospital, Third Military Medical University, Shigatse 857099, China
| | - Lingling Yang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Min Lin
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Ping Deng
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Zhiqi Yang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing 400038, China
| | - Mingyue Gong
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing 400038, China
| | - Huijie Zhang
- School of Medicine, Guangxi University, Nanning 530004, Guangxi Zhuang Autonomous Region, China
| | - Muxue Lu
- School of Medicine, Guangxi University, Nanning 530004, Guangxi Zhuang Autonomous Region, China
| | - Yanqi Li
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Peng Gao
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Yonghui Lu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Mindi He
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Lei Zhang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Huifeng Pi
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Kuan Zhang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing 400038, China
| | - Song Qin
- Department of Anatomy, Histology and Embryology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhengping Yu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Zhou Zhou
- Department of Environmental Medicine, School of Public Health, and Department of Emergency Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China; Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing 400030, China.
| | - Chunhai Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
16
|
Ben Fradj S, Nédélec E, Salvi J, Fouesnard M, Huillet M, Pallot G, Cansell C, Sanchez C, Philippe C, Gigot V, Lemoine A, Trompier D, Henry T, Petrilli V, Py BF, Guillou H, Loiseau N, Ellero-Simatos S, Nahon JL, Rovère C, Grober J, Boudry G, Douard V, Benani A. Evidence for Constitutive Microbiota-Dependent Short-Term Control of Food Intake in Mice: Is There a Link with Inflammation, Oxidative Stress, Endotoxemia, and GLP-1? Antioxid Redox Signal 2022; 37:349-369. [PMID: 35166124 DOI: 10.1089/ars.2021.0095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Aims: Although prebiotics, probiotics, and fecal transplantation can alter the sensation of hunger and/or feeding behavior, the role of the constitutive gut microbiota in the short-term regulation of food intake during normal physiology is still unclear. Results: An antibiotic-induced microbiota depletion study was designed to compare feeding behavior in conventional and microbiota-depleted mice. Tissues were sampled to characterize the time profile of microbiota-derived signals in mice during consumption of either standard or high-fat food for 1 h. Pharmacological and genetic tools were used to evaluate the contribution of postprandial endotoxemia and inflammatory responses in the short-term regulation of food intake. We observed constitutive microbial and macronutrient-dependent control of food intake at the time scale of a meal; that is, within 1 h of food introduction. Specifically, microbiota depletion increased food intake, and the microbiota-derived anorectic effect became significant during the consumption of high-fat but not standard food. This anorectic effect correlated with a specific postprandial microbial metabolic signature, and did not require postprandial endotoxemia or an NOD-, LRR-, and Pyrin domain-containing protein 3-inflammasome-mediated inflammatory response. Innovation and Conclusion: These findings show that the gut microbiota controls host appetite at the time scale of a meal under normal physiology. Interestingly, a microbiota-derived anorectic effect develops specifically with a high-fat meal, indicating that gut microbiota activity is involved in the satietogenic properties of foods. Antioxid. Redox Signal. 37, 349-369.
Collapse
Affiliation(s)
- Selma Ben Fradj
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS (UMR6265), INRAE (UMR1324), Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Emmanuelle Nédélec
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS (UMR6265), INRAE (UMR1324), Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Juliette Salvi
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS (UMR6265), INRAE (UMR1324), Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Mélanie Fouesnard
- Institut Micalis, INRAE (UMR1319), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France.,Institut NuMeCan, INRAE (UMR1341), INSERM (UMR1241), Université de Rennes 1, St-Gilles, France
| | - Marine Huillet
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse 3, INRAE (UMR1331), ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Gaëtan Pallot
- Centre de Recherche Lipides, Nutrition, Cancer, INSERM (UMR1231), Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Céline Cansell
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS (UMR7275), Université Côte d'Azur, Valbonne, France
| | - Clara Sanchez
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS (UMR7275), Université Côte d'Azur, Valbonne, France
| | - Catherine Philippe
- Institut Micalis, INRAE (UMR1319), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Vincent Gigot
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS (UMR6265), INRAE (UMR1324), Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Aleth Lemoine
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS (UMR6265), INRAE (UMR1324), Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Doriane Trompier
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS (UMR6265), INRAE (UMR1324), Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Thomas Henry
- CIRI, Centre International de Recherche en Infectiologie, Inserm (U1111), CNRS (UMR5308), ENS de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Virginie Petrilli
- Centre de Recherche en Cancérologie de Lyon, Inserm (U1052), CNRS (UMR5286), Université de Lyon 1, Lyon, France
| | - Benedicte F Py
- CIRI, Centre International de Recherche en Infectiologie, Inserm (U1111), CNRS (UMR5308), ENS de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Hervé Guillou
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse 3, INRAE (UMR1331), ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Nicolas Loiseau
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse 3, INRAE (UMR1331), ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Sandrine Ellero-Simatos
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse 3, INRAE (UMR1331), ENVT, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Jean-Louis Nahon
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS (UMR7275), Université Côte d'Azur, Valbonne, France
| | - Carole Rovère
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS (UMR7275), Université Côte d'Azur, Valbonne, France
| | - Jacques Grober
- Centre de Recherche Lipides, Nutrition, Cancer, INSERM (UMR1231), Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Gaelle Boudry
- Institut NuMeCan, INRAE (UMR1341), INSERM (UMR1241), Université de Rennes 1, St-Gilles, France
| | - Véronique Douard
- Institut Micalis, INRAE (UMR1319), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Alexandre Benani
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS (UMR6265), INRAE (UMR1324), Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
17
|
Nampoothiri S, Nogueiras R, Schwaninger M, Prevot V. Glial cells as integrators of peripheral and central signals in the regulation of energy homeostasis. Nat Metab 2022; 4:813-825. [PMID: 35879459 PMCID: PMC7613794 DOI: 10.1038/s42255-022-00610-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 06/15/2022] [Indexed: 01/03/2023]
Abstract
Communication between the periphery and the brain is key for maintaining energy homeostasis. To do so, peripheral signals from the circulation reach the brain via the circumventricular organs (CVOs), which are characterized by fenestrated vessels lacking the protective blood-brain barrier (BBB). Glial cells, by virtue of their plasticity and their ideal location at the interface of blood vessels and neurons, participate in the integration and transmission of peripheral information to neuronal networks in the brain for the neuroendocrine control of whole-body metabolism. Metabolic diseases, such as obesity and type 2 diabetes, can disrupt the brain-to-periphery communication mediated by glial cells, highlighting the relevance of these cell types in the pathophysiology of such complications. An improved understanding of how glial cells integrate and respond to metabolic and humoral signals has become a priority for the discovery of promising therapeutic strategies to treat metabolic disorders. This Review highlights the role of glial cells in the exchange of metabolic signals between the periphery and the brain that are relevant for the regulation of whole-body energy homeostasis.
Collapse
Affiliation(s)
- Sreekala Nampoothiri
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, Lille, France
| | - Ruben Nogueiras
- Universidade de Santiago de Compostela-Instituto de Investigation Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatologia de la Obesidad y Nutrition, Santiago de Compostela, Spain
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, Lille, France.
| |
Collapse
|
18
|
Mattar P, Toledo-Valenzuela L, Hernández-Cáceres MP, Peña-Oyarzún D, Morselli E, Perez-Leighton C. Integrating the effects of sucrose intake on the brain and white adipose tissue: Could autophagy be a possible link? Obesity (Silver Spring) 2022; 30:1143-1155. [PMID: 35578809 DOI: 10.1002/oby.23411] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 01/07/2022] [Accepted: 01/07/2022] [Indexed: 01/18/2023]
Abstract
Excess dietary sucrose is associated with obesity and metabolic diseases. This relationship is driven by the malfunction of several cell types and tissues critical for the regulation of energy balance, including hypothalamic neurons and white adipose tissue (WAT). However, the mechanisms behind these effects of dietary sucrose are still unclear and might be independent of increased adiposity. Accumulating evidence has indicated that dysregulation of autophagy, a fundamental process for maintenance of cellular homeostasis, alters energy metabolism in hypothalamic neurons and WAT, but whether autophagy could mediate the detrimental effects of dietary sucrose on hypothalamic neurons and WAT that contribute to weight gain is a matter of debate. In this review, we examine the hypothesis that dysregulated autophagy in hypothalamic neurons and WAT is an adiposity-independent effect of sucrose that contributes to increased body weight gain. We propose that excess dietary sucrose leads to autophagy unbalance in hypothalamic neurons and WAT, which increases caloric intake and body weight, favoring the emergence of obesity and metabolic diseases.
Collapse
Affiliation(s)
- Pamela Mattar
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Lilian Toledo-Valenzuela
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María Paz Hernández-Cáceres
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile
| | - Daniel Peña-Oyarzún
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile
- Interdisciplinary Center for Research in Territorial Health of the Aconcagua Valley (CIISTe Aconcagua, School of Medicine, Faculty of Medicine, San Felipe Campus, University of Valparaiso, Valparaíso, Chile
| | - Eugenia Morselli
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudio Perez-Leighton
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
19
|
Ameroso D, Meng A, Chen S, Felsted J, Dulla CG, Rios M. Astrocytic BDNF signaling within the ventromedial hypothalamus regulates energy homeostasis. Nat Metab 2022; 4:627-643. [PMID: 35501599 PMCID: PMC9177635 DOI: 10.1038/s42255-022-00566-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/28/2022] [Indexed: 11/12/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is essential for maintaining energy and glucose balance within the central nervous system. Because the study of its metabolic actions has been limited to effects in neuronal cells, its role in other cell types within the brain remains poorly understood. Here we show that astrocytic BDNF signaling within the ventromedial hypothalamus (VMH) modulates neuronal activity in response to changes in energy status. This occurs via the truncated TrkB.T1 receptor. Accordingly, either fasting or central BDNF depletion enhances astrocytic synaptic glutamate clearance, thereby decreasing neuronal activity in mice. Notably, selective depletion of TrkB.T1 in VMH astrocytes blunts the effects of energy status on excitatory transmission, as well as on responses to leptin, glucose and lipids. These effects are driven by increased astrocytic invasion of excitatory synapses, enhanced glutamate reuptake and decreased neuronal activity. We thus identify BDNF/TrkB.T1 signaling in VMH astrocytes as an essential mechanism that participates in energy and glucose homeostasis.
Collapse
Affiliation(s)
- Dominique Ameroso
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Alice Meng
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Stella Chen
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Jennifer Felsted
- Graduate Program in Biochemical and Molecular Nutrition, Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Chris G Dulla
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Maribel Rios
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA.
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA.
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
20
|
Ma Y, Murgia N, Liu Y, Li Z, Sirakawin C, Konovalov R, Kovzel N, Xu Y, Kang X, Tiwari A, Mwangi PM, Sun D, Erfle H, Konopka W, Lai Q, Najam SS, Vinnikov IA. Neuronal miR-29a protects from obesity in adult mice. Mol Metab 2022; 61:101507. [PMID: 35490865 PMCID: PMC9114687 DOI: 10.1016/j.molmet.2022.101507] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/12/2022] [Accepted: 04/25/2022] [Indexed: 12/30/2022] Open
Abstract
Objective Obesity, a growing threat to the modern society, represents an imbalance of metabolic queues that normally signal to the arcuate hypothalamic nucleus, a critical brain region sensing and regulating energy homeostasis. This is achieved by various neurons many of which developmentally originate from the proopiomelanocortin (POMC)-expressing lineage. Within the mature neurons originating from this lineage, we aimed to identify non-coding genes in control of metabolic function in the adulthood. Methods In this work, we used microRNA mimic delivery and POMCCre-dependent CRISPR-Cas9 knock-out strategies in young or aged mice. Importantly, we also used CRISPR guides directing suicide cleavage of Cas9 to limit the off-target effects. Results Here we found that mature neurons originating from the POMC lineage employ miR-29a to protect against insulin resistance obesity, hyperphagia, decreased energy expenditure and obesity. Moreover, we validated the miR-29 family as a prominent regulator of the PI3K-Akt-mTOR pathway. Within the latter, we identified a direct target of miR-29a-3p, Nras, which was up-regulated in those and only those mature POMCCreCas9 neurons that were effectively transduced by anti-miR-29 CRISPR-equipped construct. Moreover, POMCCre-dependent co-deletion of Nras in mature neurons attenuated miR-29 depletion-induced obesity. Conclusions Thus, the first to our knowledge case of in situ Cre-dependent CRISPR-Cas9-mediated knock-out of microRNAs in a specific hypothalamic neuronal population helped us to decipher a critical metabolic circuit in adult mice. This work significantly extends our understanding about the involvement of neuronal microRNAs in homeostatic regulation. Delivery of miR-29a-3p to the arcuate hypothalamic nucleus attenuates obesity. Knock-out of genes in mature neurons by Cre-dependent CRISPR/Cas9 technique involving Cas9-cleaving sgRNAs to limit off-target effects. Deletion of miR-29a in mature PomcCre neurons leads to early-onset insulin resistance and later to hyperphagia and decreased energy expenditure. POMCCre-restricted deletion of miR-29a causes cell-autonomous Nras up-regulation leading to obesity. POMCCre-restricted knock-out of Nras, a direct target of miR-29a-3p, attenuates obesity in mice.
Collapse
Affiliation(s)
- Yuan Ma
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Nicola Murgia
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Liu
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Zixuan Li
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Chaweewan Sirakawin
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ruslan Konovalov
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Nikolai Kovzel
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Xu
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xuejia Kang
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Anshul Tiwari
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Patrick Malonza Mwangi
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Donglei Sun
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Holger Erfle
- Advanced Biological Screening Facility, BioQuant, University of Heidelberg, Heidelberg, Germany
| | - Witold Konopka
- Laboratory of Neuroplasticity and Metabolism, Department of Life Sciences and Biotechnology, Łukasiewicz PORT Polish Center for Technology Development, Wrocław, Poland
| | - Qingxuan Lai
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Syeda Sadia Najam
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ilya A Vinnikov
- Laboratory of Molecular Neurobiology, Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Biology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
21
|
Leyrer-Jackson JM, Hood LE, Olive MF. Sex differences and the lack of effects of chemogenetic manipulation of pro-opiomelanocortin (POMC) neurons on alcohol consumption in male and female mice. Brain Res 2022; 1786:147901. [PMID: 35367433 DOI: 10.1016/j.brainres.2022.147901] [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: 12/29/2021] [Revised: 03/02/2022] [Accepted: 03/29/2022] [Indexed: 11/17/2022]
Abstract
The endogenous opioid system has been implicated in the rewarding and reinforcing effects of alcohol. Pro-opiomelanocortin (POMC) neurons located within the arcuate nucleus of the hypothalamus (ArcN) secrete multiple peptides associated with alcohol consumption, including β-endorphin (β-END), α-melanocyte stimulating hormone (α-MSH), and adrenocorticotropic hormone (ACTH). In this study, we utilized chemogenetics to bidirectionally modulate ArcN POMC neurons to determine their role in alcohol and saccharin consumption and regional levels of POMC-derived peptides. Male and female POMC-cre mice were infused with viral vectors designed for cre-dependent expression of either excitatory and inhibitory DREADDs or a control vector into the ArcN. Following recovery, animals were allowed to consume alcohol or saccharin using the drinking-in-the-dark (DID) paradigm of binge-like intake for 4 consecutive days. Prior to the final test session, animals were injected with clozapine-N-oxide (2.5 mg/kg, i.p.) for DREADD activation. Following the last DID session, animals were euthanized and the ArcN, VTA, amygdala and NAc were dissected and assessed for POMC peptide expression utilizing western blotting. We found that female mice consumed more alcohol than males during DID sessions 2-4, and that chemogenetic activation had no effect on alcohol or saccharin consumption in either sex. We found that β-END expression within the ArcN positively correlated with alcohol consumption. Given the molecular and functional heterogeneity of ArcN POMC neurons, future studies are needed to assess the effects of modulation of specific subpopulations of these neurons within the ArcN on consumption of rewarding substances such as alcohol and saccharin.
Collapse
Affiliation(s)
| | - Lauren E Hood
- Department of Psychology, Arizona State University, Tempe, AZ 85281, USA
| | - M Foster Olive
- Department of Psychology, Arizona State University, Tempe, AZ 85281, USA
| |
Collapse
|
22
|
Caron A, Jane Michael N. New Horizons: Is Obesity a Disorder of Neurotransmission? J Clin Endocrinol Metab 2021; 106:e4872-e4886. [PMID: 34117881 DOI: 10.1210/clinem/dgab421] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Indexed: 11/19/2022]
Abstract
Obesity is a disease of the nervous system. While some will view this statement as provocative, others will take it as obvious. Whatever our side is, the pharmacology tells us that targeting the nervous system works for promoting weight loss. It works, but at what cost? Is the nervous system a safe target for sustainable treatment of obesity? What have we learned-and unlearned-about the central control of energy balance in the last few years? Herein we provide a thought-provoking exploration of obesity as a disorder of neurotransmission. We discuss the state of knowledge on the brain pathways regulating energy homeostasis that are commonly targeted in anti-obesity therapy and explore how medications affecting neurotransmission such as atypical antipsychotics, antidepressants, and antihistamines relate to body weight. Our goal is to provide the endocrine community with a conceptual framework that will help expending our understanding of the pathophysiology of obesity, a disease of the nervous system.
Collapse
Affiliation(s)
- Alexandre Caron
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
- Quebec Heart and Lung Institute, Quebec City, QC, Canada
- Montreal Diabetes Research Center, Montreal, QC, Canada
| | - Natalie Jane Michael
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
- Quebec Heart and Lung Institute, Quebec City, QC, Canada
| |
Collapse
|
23
|
Órdenes P, Villar PS, Tarifeño-Saldivia E, Salgado M, Elizondo-Vega R, Araneda RC, García-Robles MA. Lactate activates hypothalamic POMC neurons by intercellular signaling. Sci Rep 2021; 11:21644. [PMID: 34737351 PMCID: PMC8569171 DOI: 10.1038/s41598-021-00947-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 10/07/2021] [Indexed: 02/03/2023] Open
Abstract
Previous studies indicate that the activity of hypothalamic POMC neurons can be regulated by glucose via intracellular mechanisms, but its regulation by lactate is poorly understood. In addition to its energetic role, lactate acts as a signaling molecule. In this study, we evaluated the function and location of the lactate receptor, hydroxycarboxylic acid receptor 1 (HCAR1). We used a conditional genetic approach to label POMC neurons and evaluated their sensitivity to lactate using patch-clamp recordings. l-Lactate and 3-chloro-5-hydroxybenzoic acid (3Cl-HBA), HCAR1 specific agonist depolarized POMC neurons and the increase in excitability was abolished by pertussis toxin (PTX), indicating the involvement of Gαi/o-protein-coupled receptors. In addition, the depolarization of a subset of POMC neurons was sensitive to α-cyano-4-hydroxycinnamate (4-CIN), a lactate transporter blocker, suggesting that the depolarization induced by l-lactate can also occur by direct intracellular action. Surprisingly, HCAR1 was not detected in POMC neurons, but instead localized in astrocytes. These results suggest a new lactate-mediated mechanism for astrocyte-neuron intercellular communication.
Collapse
Affiliation(s)
- P Órdenes
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - P S Villar
- Department of Biology Bioscience Research Bldg R-1114, University of Maryland, College Park, MD, 20742, USA
| | - E Tarifeño-Saldivia
- Gene Expression and Regulation Laboratory, Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - M Salgado
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - R Elizondo-Vega
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Ricardo C Araneda
- Department of Biology Bioscience Research Bldg R-1114, University of Maryland, College Park, MD, 20742, USA.
| | - María A García-Robles
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
| |
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
|
Astrocyte Gliotransmission in the Regulation of Systemic Metabolism. Metabolites 2021; 11:metabo11110732. [PMID: 34822390 PMCID: PMC8623475 DOI: 10.3390/metabo11110732] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 12/28/2022] Open
Abstract
Normal brain function highly relies on the appropriate functioning of astrocytes. These glial cells are strategically situated between blood vessels and neurons, provide significant substrate support to neuronal demand, and are sensitive to neuronal activity and energy-related molecules. Astrocytes respond to many metabolic conditions and regulate a wide array of physiological processes, including cerebral vascular remodeling, glucose sensing, feeding, and circadian rhythms for the control of systemic metabolism and behavior-related responses. This regulation ultimately elicits counterregulatory mechanisms in order to couple whole-body energy availability with brain function. Therefore, understanding the role of astrocyte crosstalk with neighboring cells via the release of molecules, e.g., gliotransmitters, into the parenchyma in response to metabolic and neuronal cues is of fundamental relevance to elucidate the distinct roles of these glial cells in the neuroendocrine control of metabolism. Here, we review the mechanisms underlying astrocyte-released gliotransmitters that have been reported to be crucial for maintaining homeostatic regulation of systemic metabolism.
Collapse
|
26
|
Lhomme T, Clasadonte J, Imbernon M, Fernandois D, Sauve F, Caron E, Lima N, Heras V, Martinez-Corral I, Müller-Fielitz H, Rasika S, Schwaninger M, Nogueiras R, Prevot V. Tanycytic networks mediate energy balance by feeding lactate to glucose-insensitive POMC neurons. J Clin Invest 2021; 131:e140521. [PMID: 34324439 DOI: 10.1172/jci140521] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/28/2021] [Indexed: 11/17/2022] Open
Abstract
Hypothalamic glucose sensing enables an organism to match energy expenditure and food intake to circulating levels of glucose, the main energy source of the brain. Here, we established that tanycytes of the hypothalamic arcuate nucleus, specialized glia that line the wall of the third ventricle, convert brain glucose supplies into lactate that they transmit through monocarboxylate transporters to arcuate proopiomelanocortin neurons, which integrate this signal to drive their activity and to adapt the metabolic response to meet physiological demands. Furthermore, this transmission required the formation of extensive Connexin-43 gap-junction-mediated metabolic networks by arcuate tanycytes. Selectively suppressing either tanycytic monocarboxylate transporters or gap junctions resulted in altered feeding behavior and energy metabolism. Tanycytic intercellular communication and lactate production are thus integral to the mechanism by which hypothalamic neurons that regulate energy and glucose homeostasis efficiently perceive alterations in systemic glucose levels as a function of the physiological state of the organism.
Collapse
Affiliation(s)
- Tori Lhomme
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Jerome Clasadonte
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Monica Imbernon
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Daniela Fernandois
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Florent Sauve
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Emilie Caron
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Natalia Lima
- CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Violeta Heras
- CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ines Martinez-Corral
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Helge Müller-Fielitz
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - S Rasika
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Ruben Nogueiras
- CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Vincent Prevot
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| |
Collapse
|
27
|
Folick A, Koliwad SK, Valdearcos M. Microglial Lipid Biology in the Hypothalamic Regulation of Metabolic Homeostasis. Front Endocrinol (Lausanne) 2021; 12:668396. [PMID: 34122343 PMCID: PMC8191416 DOI: 10.3389/fendo.2021.668396] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/05/2021] [Indexed: 12/18/2022] Open
Abstract
In mammals, myeloid cells help maintain the homeostasis of peripheral metabolic tissues, and their immunologic dysregulation contributes to the progression of obesity and associated metabolic disease. There is accumulating evidence that innate immune cells also serve as functional regulators within the mediobasal hypothalamus (MBH), a critical brain region controlling both energy and glucose homeostasis. Specifically, microglia, the resident parenchymal myeloid cells of the CNS, play important roles in brain physiology and pathology. Recent studies have revealed an expanding array of microglial functions beyond their established roles as immune sentinels, including roles in brain development, circuit refinement, and synaptic organization. We showed that microglia modulate MBH function by transmitting information resulting from excess nutrient consumption. For instance, microglia can sense the excessive consumption of saturated fats and instruct neurons within the MBH accordingly, leading to responsive alterations in energy balance. Interestingly, the recent emergence of high-resolution single-cell techniques has enabled specific microglial populations and phenotypes to be profiled in unprecedented detail. Such techniques have highlighted specific subsets of microglia notable for their capacity to regulate the expression of lipid metabolic genes, including lipoprotein lipase (LPL), apolipoprotein E (APOE) and Triggering Receptor Expressed on Myeloid Cells 2 (TREM2). The discovery of this transcriptional signature highlights microglial lipid metabolism as a determinant of brain health and disease pathogenesis, with intriguing implications for the treatment of brain disorders and potentially metabolic disease. Here we review our current understanding of how changes in microglial lipid metabolism could influence the hypothalamic control of systemic metabolism.
Collapse
Affiliation(s)
- Andrew Folick
- Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Suneil K. Koliwad
- Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Martin Valdearcos
- Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
28
|
González-García I, Gruber T, García-Cáceres C. Insulin action on astrocytes: From energy homeostasis to behaviour. J Neuroendocrinol 2021; 33:e12953. [PMID: 33724579 DOI: 10.1111/jne.12953] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/14/2022]
Abstract
Astrocytes are specialised glial cells that integrate distinct inputs arising from neurones, other glial cells and the microcirculation to regulate diverse aspects of brain function. A growing body of emerging evidence supports that astrocytes, similar to neurones, also play active roles in the neuroendocrine control of metabolism by responding to afferent nutritional and hormonal cues and translating these metabolic cues into neuronal inputs. Specifically, insulin action in astrocytes has received special emphasis given its newly discovered regulatory role in brain glucose uptake, which until recently was assumed to be an insulin independent process. We now know that insulin signalling in astrocytes regulates metabolic processes and behavioural responses through coupling brain glucose uptake with nutrient availability to maintain energy balance and systemic glucose homeostasis. Moreover, genetic ablation of the insulin receptor in astrocytes is associated with anxiety- and depressive-like behaviours, confirming that these glial cells are involved in the regulation of cognition and mood via insulin action. Here, we provide a comprehensive review of the most relevant findings that have been made over the course of the last few years linking insulin signalling in astrocytes with the pathogenesis of brain metabolic and neurodegenerative diseases; a still unexplored field, but with a high translational potential for developing therapies.
Collapse
Affiliation(s)
- Ismael González-García
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Tim Gruber
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Cristina García-Cáceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
29
|
Abstract
The endogenous timekeeping system evolved to anticipate the time of the day through the 24 hours cycle of the Earth's rotation. In mammals, the circadian clock governs rhythmic physiological and behavioral processes, including the daily oscillation in glucose metabolism, food intake, energy expenditure, and whole-body insulin sensitivity. The results from a series of studies have demonstrated that environmental or genetic alterations of the circadian cycle in humans and rodents are strongly associated with metabolic diseases such as obesity and type 2 diabetes. Emerging evidence suggests that astrocyte clocks have a crucial role in regulating molecular, physiological, and behavioral circadian rhythms such as glucose metabolism and insulin sensitivity. Given the concurrent high prevalence of type 2 diabetes and circadian disruption, understanding the mechanisms underlying glucose homeostasis regulation by the circadian clock and its dysregulation may improve glycemic control. In this review, we summarize the current knowledge on the tight interconnection between the timekeeping system, glucose homeostasis, and insulin sensitivity. We focus specifically on the involvement of astrocyte clocks, at the organism, cellular, and molecular levels, in the regulation of glucose metabolism.
Collapse
Affiliation(s)
- Olga Barca-Mayo
- Circadian and Glial Biology Lab, Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Miguel López
- NeurObesity Lab, Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
30
|
Haigh JL, New LE, Filippi BM. Mitochondrial Dynamics in the Brain Are Associated With Feeding, Glucose Homeostasis, and Whole-Body Metabolism. Front Endocrinol (Lausanne) 2020; 11:580879. [PMID: 33240218 PMCID: PMC7680879 DOI: 10.3389/fendo.2020.580879] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
The brain is responsible for maintaining whole-body energy homeostasis by changing energy input and availability. The hypothalamus and dorsal vagal complex (DVC) are the primary sites of metabolic control, able to sense both hormones and nutrients and adapt metabolism accordingly. The mitochondria respond to the level of nutrient availability by fusion or fission to maintain energy homeostasis; however, these processes can be disrupted by metabolic diseases including obesity and type II diabetes (T2D). Mitochondrial dynamics are crucial in the development and maintenance of obesity and T2D, playing a role in the control of glucose homeostasis and whole-body metabolism across neurons and glia in the hypothalamus and DVC.
Collapse
Affiliation(s)
| | | | - Beatrice M. Filippi
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
31
|
Cansell C, Stobbe K, Sanchez C, Le Thuc O, Mosser CA, Ben-Fradj S, Leredde J, Lebeaupin C, Debayle D, Fleuriot L, Brau F, Devaux N, Benani A, Audinat E, Blondeau N, Nahon JL, Rovère C. Dietary fat exacerbates postprandial hypothalamic inflammation involving glial fibrillary acidic protein-positive cells and microglia in male mice. Glia 2020; 69:42-60. [PMID: 32659044 DOI: 10.1002/glia.23882] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/15/2022]
Abstract
In humans, obesity is associated with brain inflammation, glial reactivity, and immune cells infiltration. Studies in rodents have shown that glial reactivity occurs within 24 hr of high-fat diet (HFD) consumption, long before obesity development, and takes place mainly in the hypothalamus (HT), a crucial brain structure for controlling body weight. Here, we sought to characterize the postprandial HT inflammatory response to 1, 3, and 6 hr of exposure to either a standard diet (SD) or HFD. HFD exposure increased gene expression of astrocyte and microglial markers (glial fibrillary acidic protein [GFAP] and Iba1, respectively) compared to SD-treated mice and induced morphological modifications of microglial cells in HT. This remodeling was associated with higher expression of inflammatory genes and differential regulation of hypothalamic neuropeptides involved in energy balance regulation. DREADD and PLX5622 technologies, used to modulate GFAP-positive or microglial cells activity, respectively, showed that both glial cell types are involved in hypothalamic postprandial inflammation, with their own specific kinetics and reactiveness to ingested foods. Thus, recurrent exacerbated postprandial inflammation in the brain might promote obesity and needs to be characterized to address this worldwide crisis.
Collapse
Affiliation(s)
- Céline Cansell
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Katharina Stobbe
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Clara Sanchez
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Ophélia Le Thuc
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Coralie-Anne Mosser
- Laboratory of Neurophysiology and New Microscopies, INSERM, Université Paris Descartes, Paris, France
| | - Selma Ben-Fradj
- CSGA, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| | - Joris Leredde
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | | | - Delphine Debayle
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Lucile Fleuriot
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Frédéric Brau
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Nadège Devaux
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Alexandre Benani
- CSGA, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| | - Etienne Audinat
- IGF, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Nicolas Blondeau
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Jean-Louis Nahon
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| | - Carole Rovère
- IPMC, CNRS, Université Côte d'Azur, IPMC, CNRS, Valbonne, France
| |
Collapse
|
32
|
Prieto R, Rosdolsky M, Hofecker V, Barrios L, Pascual JM. Craniopharyngioma treatment: an updated summary of important clinicopathological concepts. Expert Rev Endocrinol Metab 2020; 15:261-282. [PMID: 32615875 DOI: 10.1080/17446651.2020.1770081] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/13/2020] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Craniopharyngiomas (CPs) are benign histological tumors that may develop at different positions along the hypothalamic-pituitary axis. Their close, heterogenous relationship to the hypothalamus makes surgical removal challenging even though this remains the primary treatment strategy. AREAS COVERED This article presents a critical overview of the pathological and clinical concepts regarding CPs that should be considered when planning treatment. Thus, we have performed a comprehensive review of detailed CP reports published between 1839 and 2020. EXPERT OPINION CP surgery should pursue maximal tumor resection while minimizing the risk of injuring the hypothalamus. Therefore, surgical strategies should be individualized for each patient. Accurate assessment of presenting symptoms and preoperative MRI has proven useful to predict the type of CP-hypothalamus relationship that will be found during surgery. CPs with dense and extensive adhesions to the hypothalamus should be highly suspected when MRI shows the hypothalamus positioned around the mid-third of the tumor and an amputated upper portion of the pituitary stalk. Symptoms related to functional impairment of the infundibulo-tuberal area of the third ventricle floor, such as obesity/hyperphagia, Fröhlich's syndrome, diabetes insipidus, and/or somnolence, also indicate risky CP-hypothalamic adhesions. In these cases, limited tumor removal is strongly advocated followed by radiation therapy.
Collapse
Affiliation(s)
- Ruth Prieto
- Department of Neurosurgery, Puerta de Hierro University Hospital , Madrid, Spain
| | | | - Verena Hofecker
- Pathologisch-anatomische Sammlung Im Narrenturm - NHM , Vienna, Austria
| | - Laura Barrios
- Statistics Department, Computing Center, CSIC , Madrid, Spain
| | - José M Pascual
- Department of Neurosurgery, La Princesa University Hospital , Madrid, Spain
| |
Collapse
|
33
|
Abstract
Food intake and energy homeostasis determine survival of the organism and species. Information on total energy levels and metabolic state are sensed in the periphery and transmitted to the brain, where it is integrated and triggers the animal to forage, prey, and consume food. Investigating circuitry and cellular mechanisms coordinating energy balance and feeding behaviors has drawn on many state-of-the-art techniques, including gene manipulation, optogenetics, virus tracing, and single-cell sequencing. These new findings provide novel insights into how the central nervous system regulates food intake, and shed the light on potential therapeutic interventions for eating-related disorders such as obesity and anorexia.
Collapse
|